Skip to main content
Erschienen in: Journal of Neurodevelopmental Disorders 1/2019

Open Access 01.12.2019 | Review

In the line-up: deleted genes associated with DiGeorge/22q11.2 deletion syndrome: are they all suspects?

verfasst von: Zahra Motahari, Sally Ann Moody, Thomas Michael Maynard, Anthony-Samuel LaMantia

Erschienen in: Journal of Neurodevelopmental Disorders | Ausgabe 1/2019

Abstract

Background

22q11.2 deletion syndrome (22q11DS), a copy number variation (CNV) disorder, occurs in approximately 1:4000 live births due to a heterozygous microdeletion at position 11.2 (proximal) on the q arm of human chromosome 22 (hChr22) (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011). This disorder was known as DiGeorge syndrome, Velo-cardio-facial syndrome (VCFS) or conotruncal anomaly face syndrome (CTAF) based upon diagnostic cardiovascular, pharyngeal, and craniofacial anomalies (McDonald-McGinn and Sullivan, Medicine 90:1-18, 2011; Burn et al., J Med Genet 30:822-4, 1993) before this phenotypic spectrum was associated with 22q11.2 CNVs. Subsequently, 22q11.2 deletion emerged as a major genomic lesion associated with vulnerability for several clinically defined behavioral deficits common to a number of neurodevelopmental disorders (Fernandez et al., Principles of Developmental Genetics, 2015; Robin and Shprintzen, J Pediatr 147:90-6, 2005; Schneider et al., Am J Psychiatry 171:627-39, 2014).

Results

The mechanistic relationships between heterozygously deleted 22q11.2 genes and 22q11DS phenotypes are still unknown. We assembled a comprehensive “line-up” of the 36 protein coding loci in the 1.5 Mb minimal critical deleted region on hChr22q11.2, plus 20 protein coding loci in the distal 1.5 Mb that defines the 3 Mb typical 22q11DS deletion. We categorized candidates based upon apparent primary cell biological functions. We analyzed 41 of these genes that encode known proteins to determine whether haploinsufficiency of any single 22q11.2 gene—a one gene to one phenotype correspondence due to heterozygous deletion restricted to that locus—versus complex multigenic interactions can account for single or multiple 22q11DS phenotypes.

Conclusions

Our 22q11.2 functional genomic assessment does not support current theories of single gene haploinsufficiency for one or all 22q11DS phenotypes. Shared molecular functions, convergence on fundamental cell biological processes, and related consequences of individual 22q11.2 genes point to a matrix of multigenic interactions due to diminished 22q11.2 gene dosage. These interactions target fundamental cellular mechanisms essential for development, maturation, or homeostasis at subsets of 22q11DS phenotypic sites.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s11689-019-9267-z) contains supplementary material, which is available to authorized users.
Abkürzungen
22q11DS
22q11.2 deletion syndrome
Ac-LDL
Acetylated low-density lipoprotein
ADHD
Attention deficit hyperactivity disorder
ASD
Autistic spectrum disorders
BTB
Bric-a-brac-Tramtrack-Broad complex
CAT
Cationic amino acid transporters
CNs
Cranial nerves
CNV
Copy number variation
CP
Cortical plate
CTAF
Conotruncal anomaly face syndrome
Dmd1
Dystrophin
Dpp
Decapentaplegic
EGF
Epidermal growth factor
ER
Endoplasmic reticulum
FGF
Fibroblast growth factor
GEFs
Guanine-nucleotide exchange factors
GPI
Glycosylphosphatidylinositol
hChr22
Human chromosome 22
IPN
Interpeduncular nucleus
iPSCs
Induced pluripotent stem cells
IZ
Intermediate zone
Iα/β
Importins
KRAB
Kruppel-associated box
LCR
Low copy number repeats
NES
Nuclear export signal
NLS
Nuclear localization signal
PI3
Phosphoinositide 3-kinase
PLC
Phospholipase C
PPI
Pre-pulse inhibition
PtdIns4P
Phosphatidylinositol 4-phosphate
PTEN
Phosphatase and tensin homolog
RA
Retinoic acid
SCZ
Schizophrenia
snoRNA
Small nucleolar RNAs
SP
Subplate
Utrn
Utrophin
VCFS
Velo-cardio-facial syndrome
VZ
Ventricular zone
Xpo1
Exportin

Background

The frequency of 22q11.2 deletion—the chromosomal/copy number variant (CNV) that results in most cases of DiGeorge/22q11DS [1, 2]—reflects the unusual genomic architecture of human chromosome 22 (hChr22), position q11.2. At least four repetitive DNA “cassettes” known as low copy-number repeats (LCRs): LCR A, B, C, and D; define the region (Fig. 1a) [6]. These LCRs, due to substantial sequence similarity, facilitate non-allelic homologous meiotic recombination, or “deletion by crossing over of repetitive DNA” [7], resulting in unbalanced translocation, deletions, or duplication [8]. More than 85% of recombination occurs between LCR A and LCR D resulting in a 3 Mb “typical” deletion. The smaller 1.5 Mb “minimal critical” deletion, occurs between LCR A and LCR B in ∼ 10% of affected individuals [6, 8]. Deleted individuals have complex and variable phenotypes including developmental delay, congenital heart defects, craniofacial anomalies such as cleft palate and retrognathia, musculoskeletal anomalies, eye anomalies, hearing loss, absent or small thymus and parathyroid glands, hypocalcemia, compromised immune system, feeding difficulties, and seizures. In addition to these physical manifestations, 22q11.2 deletion syndrome (22q11DS) is associated with behavioral and psychiatric complications such as schizophrenia (SCZ) autistic spectrum disorders (ASD), attention deficit hyperactivity disorder (ADHD), anxiety disorder, as well as intellectual and learning disabilities [35]. Clearly, the range of genomic lesions, numbers of genes, and spectrum of phenotypes that define 22q11 CNV disorders defies straightforward explanations of genotype to phenotype correlations. The essential question remains: how CNVs of one, many, or all 22q11.2 genes disrupts developmental and homeostatic mechanisms and complicates the lives of children and adults with 22q11DS.
Individuals with the A to B deletion (we use this nomenclature to identify heterozygous elimination of 22q11.2 genes between LCRs A and B) have the full spectrum of phenotypes seen also with the typical A to D deletion (i.e., elimination of genes between LCRs A and D), suggesting that key 22q11DS phenotypes are largely due to diminished A to B gene dosage [911]. Microcephaly and ocular anomalies occur in about 50% of individuals with A to B or A to D deletions compared to 7% in B to D or C to D deleted individuals, and cardiovascular defects also appear to be > 3 times more frequent in A to B or A to D deletions [12]. Nevertheless, deletions in distal regions, B to D or C to D, have been associated with cardiac developmental anomalies similar to those associated with A to B or A to D deletions, albeit with lower frequency [13]. Furthermore, the presence and severity of phenotypes among individuals carrying the A to B deletion [14] varies significantly, even in siblings who inherit the same deletion from a 22q11.2-deleted parent [15]. In the context of these complex associations between penetrance and severity, it is unlikely that haploinsufficiency of TBX1 (a 22q11.2 A to B gene proposed to explain cardiovascular malformation) [16] or any other single 22q11.2 gene, accounts for the broad range of heart defects in 22q11.2-deleted individuals. This suggests that stochastic modifying interactions between genes in the A to B region, as well as unrecognized modifying functions of B to D genes, or convergence of subsets of 22q11.2 deleted genes on specific cellular mechanisms play a role in 22q11DS pathogenesis. We will evaluate this question based upon assessment of apparent obligate cellular function (that disrupted by full loss-of-function) of each of the 41 characterized proteins between LCRs A and D, based upon genetic as well as cell biological analyses in human and diverse model systems or organisms.
Phenotypic variability in 22q11DS extends to behavioral diagnoses associated with the syndrome. Several commonly diagnosed disorders, including ASD, are apparently more frequent in A to B deleted individuals [12, 17]. Furthermore, several genes from A to D including COMT, PRODH, GNB1L, TBX1, SEPT5/GP1BB, ZDHHC8, PI4KA, and ARVCF have been individually associated with SCZ, ASD, ADHD, and other disorders frequently diagnosed in individuals with 22q11DS [1820]. Recent population-based exome analysis, however, supports polygenic inheritance of broader, clinically defined behavioral disorders. In ASD, aside from 22q11.2, five additional CNV loci: 1q21.1, 3q29, 7q11.23, 16p11.2, and 15q11.2–13, as well as multiple de novo gene variants have been identified as risk factors [21]. Similarly, SCZ is associated with multiple, rare loss and gain of function mutations in genes that encode calcium channels, postsynaptic cytoskeleton-associated scaffold proteins, and cell adhesion/signaling molecules [22, 23]. Whole exome sequence analyses in individuals with non-syndromic congenital heart defects also fail to support robust individual gene/phenotype correlations [24]. It is thus unlikely that developmental phenotypes in individuals with 22q11.2 CNVs—including commonly diagnosed behavioral deficits associated with neurodevelopmental disorders—arise from the consequences of altered dosage of one single gene within the A to D region. Indeed, single gene explanation for individual phenotypes or complete phenotypic spectra are unlikely for most CNV syndromes, including fairly common deletions and duplications at 7q11.23 (Williams Syndrome), 15q11.2 (Prader-Willi Angelman Syndrome), 16p11.2 (Autism Susceptibility), and even whole or partial chromosomal anomalies like trisomy 21 (Down syndrome). Thus, a reassessment of the known protein coding genes at 22q11.2 between LCRs A and D, and their causal relationship to 22q11DS, based upon a thorough review of molecular and cellular mechanisms through which each gene influences development or ongoing physiological function, is necessary, timely, and may have broader resonance for understanding several dosage-related, CNV-associated multigenic neurodevelopmental syndromes.

Main text

The 22Q11.2 gene “line-up”: diverse genes on hChr 22q11.2 from A to D

The genes deleted in 22q11DS are conserved across mammals with only modest rearrangements in the human versus mouse minimal deleted regions [3, 25, 26]. Thus, the mouse has emerged as the most common, accessible, genomically valid model for mammalian 22q11.2 gene function [26]. Drosophila and Caenorhabditis elegans as well as several non-mammalian vertebrates including zebrafish, frog, and chicken also have orthologues of most 22q11.2 genes [3, 25]. There are 154 GenBank entries in the 3 Mb deleted region of hChr22 (LCR A to LCR D; annotation release 108), of which only 56 are predicted to encode proteins: 41 are characterized, while 15 have not been evaluated in any tissue (Additional file 1: Figure. S1). The A to B deletion includes 36 (28 characterized) of the 56 apparent protein-coding loci, and the additional 20 (13 characterized) are found in the B to D region. The A to D interval also contains seven microRNAs, 38 non-coding RNAs, and 53 pseudogenes (Additional file 1: Figure. S1). To better understand the functional and phenotypic consequences of 22q11.2 copy number variation, we divided the 41 A to D genes that encode known proteins into ten categories based upon established or inferred primary cellular functions or membership in larger gene families (Fig. 1b). Our criteria included shared functional domains or organelle localization sequences in each of the proteins, common subcellular localization in multiple cell types, and literature reports of apparent obligate functions of each gene. We then re-evaluated available data on specific functions of these protein-coding genes within these categories with a focus on potential contributions to four major 22q11DS phenotypes: heart, face, immune, and brain/behavioral anomalies based upon on the cellular mechanism-based categories.

Chromatin modification/DNA replication genes

HIRA Histone Cell Cycle Regulator (a.k.a. DGCR1 or TUPLE1; A to B) encodes a histone chaperone responsible for replication-independent chromatin incorporation of histone variant H3.3 [27]. HIRA, conserved across eukaryotes, is implicated in transcriptional silencing [2833]. Its N-terminal domain also functions as a transcriptional activator marking active genes and enhancers [3437], and potentially involved in transcriptional elongation and recovery after DNA damage [38, 39]. Post-translational modification of HIRA is crucial for MyoD activation during muscle cell differentiation [40]. HIRA has two nuclear localization domains and seven WD40 repeats for protein-protein interactions [41, 42]. Among potential interactors, HIRA complexes with Pax3 [43], critical for neural, cardiac, craniofacial, thymic, thyroid, and parathyroid development [44]. In Xenopus, hira downregulation impairs gastrulation [45]. In mouse and chicken, Hira is detected in neural ectoderm, premigratory and migratory neural crest, limb buds, pharyngeal arches, and heart (Fig. 1c) [42, 46]. HIRA depletion in chicks increases persistent truncus arteriosus, a key 22q11DS phenotype [47]. Conditional Hira deletion in mouse cardiac myocytes alters gene expression, causes pathology and hypertrophy, but does not perturb cardiac morphogenesis [48]. Hira−/− mice, however, have severe phenotypes including gastrulation and neurulation defects, growth retardation, and aberrant cardiac development leading to embryonic lethality [41]. No phenotypes have been reported in heterozygous embryos, neonates, or adults.
CDC45 Cell Division Cycle 45 (A to B) is required for initiation, elongation, and coordination of eukaryotic chromosomal DNA replication [4954]. Data from fission yeast, Xenopus egg extracts, and human cells indicate that CDC45 is a limiting factor for replication initiation [5557]. Cdc45 is expressed in embryonic brain, pharyngeal arches, thymus and thyroid—where neural crest augments endodermal derivatives (Fig. 1d) [5860]. Cdc45−/− mice have impaired inner cell mass proliferation and die shortly after implantation [61]. Heterozygotes, however, have no apparent phenotypes. Human CDC45 loss-of-function mutations are linked to craniosynostosis (premature cranial suture closure) and Meier-Gorlin Syndrome, a rare autosomal recessive disorder characterized by growth retardation, microcephaly, small ears and jaws, palatal anomalies, hearing, and vision loss [62], which are very similar to 22q11DS phenotypes [4].

Perspective: chromatin modification/DNA replication genes

Apparently, the chromatin remodeling/DNA replication genes HIRA and CDC45, in the A to B region, are necessary for embryonic survival and modulate neural, cardiovascular, and craniofacial development. Loss of function of either gene in mice results in embryonic lethality; however, heterozygotes have no reported anomalies. The appearance of this gene class, chromatin modifiers, in only the A to B region elevates their status as likely contributors to key 22q11DS phenotypes. Phenotypic severity, however, may depend upon diminished dosage of other A to B genes since neither Hira nor Cdc45 heterozygous deletion in mouse yields detectable phenotypes.

Signaling/adaptor factors

GNB1L G-Protein Subunit Beta (a.k.a. Wdvcf; A to B) encodes a G-protein/WD40 repeat protein of unknown function [63]. It is highly expressed in mouse forebrain, midbrain, and hindbrain (Fig. 1c) [64]. Homozygous mutation is embryonic lethal, and heterozygotes have pre-pulse inhibition (PPI) deficits, a potentially SCZ-related behavioral phenotype in animal models [64].
CRKL Chicken tumor virus number 10 regulator of kinase (B to D) encodes an adapter protein with multiple SH2 and SH3 protein-protein interaction domains [65]. CRK proteins transmit extracellular signals by physically bridging tyrosine-phosphorylated proteins to mediators like Abl tyrosine kinase and guanine-nucleotide exchange factors (GEFs), to influence cell proliferation, differentiation, migration, adhesion, and immune responses [66]. Crkl binds to both phosphorylated Dab1, a mediator of Reelin signaling, and C3G, a Rap1 GEF, to coordinate neuronal migration by cytoskeletal regulation [6769]. Crkl is expressed at high levels in pharyngeal arches, neural crest, and brain, and homozygous deletion compromises neural crest derivatives including cranial ganglia, aortic arch arteries, thymus, parathyroid glands, and craniofacial structures, leading to late-gestation lethality [70]. Homozygous loss-of-function in Snoopy, a point Crkl mutation [71], also results in craniofacial anomalies, pharyngeal occlusion, and holoprosencephaly. Snoopy or Crkl+/−:Tbx1+/− compound heterozygotes (Tbx1, an A to B gene, see below) have increased retinoic acid (RA) signaling, also seen in mouse embryos with an A to B deletion (LgDel) [7174]. Crkl may interact with other inductive signaling pathways; fibroblast growth factor (FGF) receptors directly bind to the Ckrl SH2 domain [75]. In 22q11DS, diminished CRKL dosage due to B to D deletion is associated with impaired T cell and natural killer cell function [76, 77], consistent with CRKL regulation of immune cells [7880].
SCARF2 Scavenger Receptor Class F Member 2 (a.k.a. SREC-II; B to D) encodes a Ca++ binding protein that interacts with small molecule ligands such as acetylated low-density lipoprotein (Ac-LDL) [81]. The SCARF2 extracellular domain has multiple epidermal growth factor (EGF)-like repeats and several positively charged residues. Its intracellular domain contains 13 potential phosphorylation sites suggesting a role in cell signaling [81]. Scarf2 is expressed in neonatal and adult mouse skin, tongue, oral epithelia, and medullary thymus [82]. Its obligate functions in the developing or adult mouse remain unclear; there are no reports of Scarf2 mutations. SCARF2 mutations have been linked to Van Den Ende-Gupta syndrome, an extremely rare autosomal recessive disorder characterized by craniofacial anomalies: narrow eye openings, maxillary hypoplasia, flat/wide nasal bridge, everted lower lip, palatal disruptions, prominent ears, skeletal anomalies: scoliosis, long, slender foot and hand bones, mild bowing of long bones, respiratory difficulty due to laryngeal deficits, and cerebellar hyperplasia [8385]. These overlap with phenotypes seen in 22q11DS [4].
PI4KA Phosphatidylinositol 4-Kinase Alpha (a.k.a. PI4KIIIα; B to D) catalyzes synthesis of phosphatidylinositol 4-phosphate (PtdIns4P), an essential precursor of PtdIns (4,5) P2 and PtdIns (3,4,5) P3 that mediate receptor-activated phospholipase C (PLC) and phosphoinositide 3-kinase (PI3) signaling [86]. The yeast PI4KA orthologue, STT4, is required for cell wall integrity and actin cytoskeleton organization [87]. PI4KA is found in endoplasmic reticulum (ER), nucleoli, and pericentriolar regions, coincident with the Golgi complex [8890]. Membrane proteins TTC7 and EFR3 recruit PI4KA to the plasma membrane to help maintain protein and lipid composition [91, 92]. In zebrafish, pi4ka loss-of-function disrupts brain, heart, and trunk development perhaps due to impaired FGF signaling [93]. PI4KA is expressed in fetal and adult rodent and human brain [94, 95]. Conditional Pi4ka inactivation causes lethal gastrointestinal mucosal degeneration, but no obvious neural anomalies [96, 97]. Biallelic PI4KA mutation, however, is associated with cerebellar hypoplasia and polymicrogyria [98]. PI4KA is also recognized as a SCZ-susceptibility gene in adults with 22q11DS [19, 99].

Perspective: signalling/adaptor genes

The A to B signaling/adaptor gene, GNB1L, may contribute to 22q11DS phenotypes, especially in the brain based upon behavioral evidence; however, current data are inconclusive. The B to D signaling/adaptor genes, CKRL, SCARF2, and PI4KA, may modulate craniofacial, cardiac, or cerebral cortical differentiation. Thus, while heterozygous CRKL, SCARF2, or PI4KA deletion may not be necessary for core 22q11DS phenotypes, diminished dosage of one or more of these genes in individuals with the A to D deletion may enhance heart, face, or brain phenotypes, yielding more severe impairments or introducing greater phenotypic variation.

Cell-cell adhesion genes

ARVCF Armadillo Repeat Gene Deleted in Velo-cardio-facial syndrome (A to B) is a member of the Catenin family, which includes α- and β-catenin, plakoglobin, and p120-related proteins [100, 101]. Catenins are critical for adherens junction assembly to facilitate extracellular and intracellular communication. ARVCF protein contains an N-terminus coiled-coil domain, 10 armadillo repeats, and a C-terminal PDZ motif that binds the scaffold protein ERBIN and tight junction proteins ZO-1 and ZO-2 [102104]. When bound, ARVCF enhances the stability of classical cadherins by reducing their endocytosis [105, 106]. When not bound to cadherin, ARVCF may regulate the cytoskeleton by interacting with small-GTPases such as Rho and Rac [107, 108]. Like most catenins, ARVCF also translocates to the nucleus where it may be involved in alternative mRNA splicing [109, 110]. ARVCF is expressed in the human forebrain ganglionic eminences; mouse Arvcf is expressed in the brainstem, hippocampus, cerebral cortex, and heart (Fig. 1c) [58, 111]. Arvcf depletion in Xenopus leads to defects in craniofacial skeleton and aortic arches (also observed in Tbx1-deficient tadpoles; see below) [112, 113]. The migration of cranial neural crest is not affected by knocking down of either arvcf or tbx1; however, their double depletion causes delayed migration suggesting these genes act cooperatively [113]. There are no reported loss-of-function Arvcf alleles; however, loss of function of p120 catenin results in substantially diminished embryonic Arvcf expression, in concert with disrupted renal differentiation [114]. ARVCF mutations and their related phenotypes do not include structural brain anomalies; however, they have been associated with increased susceptibility to SCZ [115, 116].
CLDN5 Claudin5, (a.k.a. TMVCF; A to B) is a member of the Claudin family, major structural components of tight junctions that are crucial for cell–cell contacts and paracellular permeability, and establishing apical to basolateral intra-membrane borders [117119]. Cytokines, growth factors, and cadherins regulate claudins [120, 121]. All family members have similar predicted folding and high sequence homology in the first and fourth transmembrane domains and extracellular loops [118], and a C terminus PDZ motif that binds to PDZ peripheral membrane proteins, including ZO-1, -2, and -3, which in turn interact with the actin cytoskeleton to link tight junctions with signal transduction proteins [122, 123]. Cldn5 is expressed ubiquitously [124, 125]. In Xenopus, cldn5 is strongly expressed in cardiac primordia and its depletion leads to heart malformations [126] and in zebrafish, cldn5 is required for heart regeneration [127]. Cldn5−/− mice, however, do not have cardiovascular malformations. Nevertheless, Cldn5 may modify cardiovascular development via interactions with additional myogenic genes [128, 129]. In the Dystrophin−/− (Dmd1)/Utrophin−/− (Utrn) cardiomyopathy model, Cldn5 mRNA and protein levels decline [129]; in contrast, Cldn5 overexpression diminishes cardiomyopathy in Dmd1−/−:Utrn−/− mice [128]. Cldn5 also is expressed in cerebral microvascular endothelial cells, perhaps regulating blood-brain barrier permeability [124, 130]. Cldn5−/− mice develop normally but die shortly after birth [130]. There is no evidence for disrupted tight junction structure; however, small molecule paracellular permeability increases selectively in the mutant brain suggesting that lack of Cldn5 alters junctional function [130]. CLDN5 is reduced in human end-stage cardiomyopathy [131, 132]. In addition, CLDN5 is implicated in stem cell self-renewal. ZO-1, CLDN1, CLDN3, and CLDN5 are highly expressed in human neural stem cells, but significantly down-regulated during neuronal differentiation [133].
DGCR2 DiGeorge Syndrome Critical Region Gene 2 (a.k.a. SEZ12; A to B) encodes a novel putative transmembrane adhesion receptor [134] expressed in the developing and adult mouse brain (Fig. 1c) [58]. Recent studies support a role for Dgcr2 in cortical projection neuron migration and differentiation, apparently as part of a complex that regulates Reelin-dependent phosphorylation of Dab1, Akt, Erk1/2 and subsequent signaling targets [135]. Dgcr2 deficient mice have gait abnormalities, reduced movement, and impaired motor coordination possibly due to loss of cerebellar Purkinje cells [136]. Finally, a DGCR2 missense mutation has been associated with non-syndromic SCZ [137], reinforcing a potential contribution to neural development or function.

Perspective: cell-cell adhesion genes

The A to B cell-cell adhesion genes, ARVCF and CLDN5, belong to key junctional complex gene families: catenin/cadherins and claudin/ZO-1, respectively. Based upon their A to B location and essential roles in adhesion, cell-cell interactions, morphogenesis, and maintenance of tissue integrity—all compromised in 22q11DS—they are likely, but as yet minimally investigated suspects for core phenotypes. Their fundamental functions assessed in vitro or by genetic manipulation in frog, zebrafish, and mouse are consistent with contributions to cardiac and neural anomalies. DGCR2, a proposed A to B SCZ vulnerability gene, is an apparent adhesion receptor involved in cortical neuronal migration. Diminished dosage of A to B adhesion genes may converge on broader signaling networks at 22q11DS phenotypic sites, or each gene may independently compromise development, differentiation, or adult function. Given the morphogenetic consequences of disrupted adhesion, the contributions of A to B cell adhesion gene dosage in 22q11DS pathogenesis merit additional genetic and functional analysis.

Protein trafficking

SEPT5 Septin 5 (a.k.a. CDCREL1 or PNUTL1; A to B) is a member of the highly conserved Septin family of GTP-binding proteins that act as dynamic, multifunctional protein scaffolds. First discovered as essential for cytokinesis in yeast, Septins are found in most eukaryotes except plants [138, 139]. They are found in protein complexes that regulate membrane dynamics, vesicle trafficking, cell cycle, cytokinesis, cytoskeletal reorganization, cell polarity, oncogenesis, and apoptosis [140]. SEPT5, expressed predominantly in neurons, was first isolated as a constituent of a synaptophysin-associated synaptic vesicle protein complex (Fig. 1c, d) [141, 142]. It localizes to presynaptic terminals, interacts with the SNARE protein Syntaxin, and negatively regulates exocytosis and neurotransmitter release [141, 143, 144]. Moreover, in mouse, Sept5 in the auditory calices of Held regulates the coupling of Ca++ influx to presynaptic neurotransmitter release, and its overexpression inhibits dopamine release in transfected neurons in vitro [145, 146]. Nevertheless, synaptic transmission is not disrupted in Sept5−/− or Sept3−/−/Sept5−/− mice, suggesting compensation by other family members [147, 148]. In mouse and rat, altered Sept5 levels are linked to defective social interactions and cognitive impairment [149152]. In addition, homozygous deletion of SEPT5 and the adjacent GP1BB has been associated with cortical dysplasia with polymicrogyria, developmental delay, platelet secretion defects, social/emotional, and language/speech deficits [153].
CLTCL1 Clathrin Heavy Chain Like 1 (a.k.a. CHC22; A to B) is a member of the CLATHRIN gene family crucial for intracellular endosome trafficking [154, 155]. Its potential function has been difficult to assess—it does not have a murine orthologue [156]. CLTCL1 is expressed in all cell types; however, levels differ between tissues and developmental stages [157, 158]. It is expressed maximally during human prenatal brain development and declines significantly by early childhood—correlated with critical periods for neural circuit differentiation [159]. CLTCL1 homozygous missense mutations are associated with a recessive disorder characterized by touch and pain insensitivity and severe intellectual disability [159]. In a human neural crest-derived cell line—relevant because nociceptive (pain) neurons are derived from the neural crest [160]—CLTCL1 downregulation induces neuronal differentiation and facilitates RA-mediated neurite outgrowth [159]. CLTCL1 is highly expressed in skeletal muscle where it modulates glucose transporter 4 intracellular trafficking [157], myogenesis, and repair [161, 162]. Neuronal and myogenic activities implicate CLTCL1 as a candidate for 22q11DS dosage-dependent phenotypes. The lack of an animal model to test hypotheses of organismal function complicates further assessment of CLTCL1’s potential contributions to the 22q11DS phenotypic spectrum.
RANBP1 RAN Binding Protein 1 (A to B) regulates the multi-protein “RAN complex” [163, 164]. The Ran complex includes the small G protein Ran, which when bound to GDP scaffolds other proteins including importins (Iα/β) that bind nuclear transport cargos (Fig. 2a). In the nucleus, the guanine exchange factor RCC1 replaces GDP bound to RAN with GTP, establishing new exportin (Xpo1) complexes that scaffold outgoing cargo. RANBP1 binds to the RAN complex as cargos exit the nucleus, displacing and releasing them into the cytoplasm and facilitating RanGAP GTPase activity [165]. RANBP1 also modulates mitotic spindle formation [166], nuclear envelope disassembly/reassembly [167], and primary cilia protein trafficking [168]. Ranbp1 is expressed in neural crest and neocortical ventricular precursors at mid-gestation (Fig. 1c, d) [169, 170]. Ranbp1−/− mice are not viable [170]. Over 50% of these embryos are exencephalic. Non-exencephalic homozygotes are smaller than WT littermates (Fig. 2b) with craniofacial and ocular anomalies (Fig. 2c). Ranbp1 remains highly expressed in cerebral cortical ventricular and subventricular proliferative zones [58, 60, 170]. Non-exencephalic Ranbp1−/− fetuses are microcephalic (Fig. 2c) with diminished cortical thickness (Fig. 2d, e). RANBP1 haploinsufficiency has not been linked to single 22q11DS phenotypes; polymorphisms are associated with SCZ-related eye tracking anomalies [171] and reduced cortical expression is linked to disrupted metabotropic glutamate receptor signaling in ASD [172].
SNAP29 Synaptosome Associated Protein 29 kDa (B to D) belongs to the SNAP25 gene family encoding T-SNAREs that mediate vesicle/target membrane fusion [173]. SNAP29 co-localizes with endosomes, lysosomes, and Golgi in non-neuronal cells and influences protein trafficking, endocytosis, exocytosis, and phagocytosis [174178]. Synaptic SNAP29 modulates neurotransmitter release by inhibiting SNARE complex disassembly [179, 180]. Snap29 also interacts with GTPase Rab3A in myelinating glia and is implicated in surface-directed myelin proteolipid protein trafficking [181]. SNAP29 loss-of-function, independent of 22q11DS, is linked to CEDNIK: Cerebral Dysgenesis, Neuropathy, Ichthyosis, and Keratoderma [182, 183], characterized by psychomotor retardation, thick, scaly skin, facial anomalies, sensorineural deafness, microcephaly, corpus callosum dysgenesis, and cortical dysplasia. Parallel brain and epidermal phenotypes in constitutive and conditional Snap2−/− mice [184] reinforces its potentially causal role in CEDNIK. Its contributions to 22q11DS phenotypes in typically deleted individuals (A to D), however, remain unanalyzed.

Perspective: protein trafficking genes

Disrupted protein trafficking due to diminished 22q11.2 gene dosage could have significant effects on signaling during development, differentiation, or in the mature nervous system and other 22q11DS phenotypic sites. CLCTL1, RANBP1 (A to B), and SNAP29 (B to D) are required for early brain development; SEPT5 (A to B) is not. Instead, in mice, Sept5 may modulate circuit development or neuronal signaling for cognitive and social behaviors. Evidence for Ranbp1-dependent regulation of neural and craniofacial differentiation is fairly strong. It is unclear, however, how diminished RANBP1 dosage interacts with other A to B or B to D genes to yield 22q11DS phenotypes. SNAP29 may modulate A to B dependent craniofacial development due to its activity in neural crest, or dosage sensitivity in non-neural crest cells that contribute to facial structures. Accordingly, based upon obligate and disease-related functions, SNAP29 may act as a modifier for some 22q11DS phenotypes.

mRNA/miRNA biogenesis and processing genes

DGCR14 DiGeorge Critical Region Gene 14 (a.k.a. ESS2 or DGSI; A to B) encodes a nuclear protein with two N-terminal coiled-coil domains [185] and is conserved across eukaryotes from yeast to human [186189]. In mice, Dgcr14 is expressed in the pharyngeal arches and neural tube [186]. Despite its expression at key sites, the consequences of loss-of-function mutations in this gene have not been reported in mice or any other model species. Dgcr14 expression has been assessed in the context of loss of function of another A to B gene, Gsc2 (see below), whose expression is limited to the interpeduncular nucleus (IPN)—a brain region that modulates dopaminergic neurotransmission, sleep, and eye movement. Dgcr14 expression, also enhanced in the IPN, is diminished following Gsc2 deletion [190]. The significance of this observation for Dgcr14 function or that of Gsc2 in the IPN, however, remains unexplored. A recent large-scale protein interactome study identified DGCR14 as a non-core component of the spliceosome C complex, suggesting a role in mRNA processing [191193]. Indeed, Dgcr14 apparently regulates IL17a transcription during TH17 cell differentiation in vitro [194], perhaps relevant for immune dysfunction in 22q11DS. In human, DGCR14 is expressed in the heart, brain, and skeletal muscle [195]. The exact function of DGCR14 in human development or mature function, however, remains uncertain.
DGCR8 DiGeorge Critical Region Gene 8 (a.k.a. Pasha; A to B) encodes a heme-binding protein known as Pasha in flies and nematodes [196]. DGCR8 is localized to the nucleus as part of the Drosha/RNaseIII microprocessor complex that processes primary microRNAs (pri-miRNA) into precursor miRNA (pre-miRNA) in the first step of miRNA maturation [196, 197]. DGCR8 also has a microprocessor-independent role in small nucleolar RNAs (snoRNAs) and telomerase RNAs biogenesis [198]. DGCR8 is required for normal development of the nervous system [199, 200]. In Drosophila, pasha influences dendrite targeting and axon terminal arborization [201, 202]. In mice, Dgcr8 is highly expressed in the brain (Fig. 1c) and loss-of-function results in pre-implantation lethality [200]. Dgcr8 heterozygous deletion, however, results in working memory deficits and impaired short-term synaptic plasticity due to defective dendritic spines and complexity [200, 203]. Furthermore, Dgcr8 targeted deletion in pyramidal neurons alters synaptic transmission [204, 205]. Dgcr8 deficiency also has been suggested to underlie aberrant cortical interneuron migration caused by disrupted Cxcr4/Cxcl12 signaling [206, 207]. DGCR8 may modulate gene expression underlying establishment and maintenance of peripheral myelination [208]. Targeted Dgcr8 loss-of-function in cardiac neural crest leads to persistent truncus arteriosus, ventricular septal defects [209], cardiomyocyte-related cardiomyopathy, and premature lethality [210].

Perspective: mRNA/miRNA biogenesis and processing genes

The A to B location of miRNA biogenesis 22q11.2 genes DGCR14 and DGCR8, acknowledged functional importance of this miRNA regulation of development and homeostasis, and requirement for DGCR14 and DGCR8 for nervous system and heart development establishes these two genes as prime suspects for understanding 22q11DS phenotypes and their variability. A circumstantial case can be made for DGCR14 function in the nervous and immune systems; however, there is still surprisingly little known about indispensable functions of this gene. The contributions of DGCR8 to 22q11DS are beyond doubt. Nevertheless, Dgcr8 heterozygous phenotypes and those of mice with full deletion of the A to B orthologue are not identical [26]. Thus, key DGCR8 functions remain to be explored further in the context of diminished dosage of additional 22q11.2 A to B as well as B to D genes.

Transcription factors

TBX1 (A to B) is a member of the “T-box” family of transcription factors. Although TBX1 is neither a strong transcriptional activator nor a strong repressor, it regulates a large number of genes through epigenetic modifications [211]. Tbx1 is expressed in pharyngeal arch endoderm and mesoderm, an expression pattern conserved in mice, chickens, Xenopus, and zebrafish [212214]. A great deal of insight—and speculation—has arisen from analyses of Tbx1 loss-and gain-of-function mutants in a variety of model species. In zebrafish, the tbx1 loss-of-function van gogh mutation results in ear, thymus, and pharyngeal arch defects [214]. In Xenopus, its loss-of-function disrupts head, pharyngeal apparatus, and heart development [212, 215]. Tbx1 heterozygous mutant mice have mild non-lethal phenotypes; homozygous null mutants, however, die at birth with cardiac outflow tract anomalies, craniofacial defects, cleft palate, and severe thymus and parathyroid abnormalities [216218]. In mouse models, the narrowing (stenosis) of the fourth pharyngeal arch artery that develops into the central portion of the aortic arch is the most prominent cardiovascular phenotype (Fig. 3a–c) [74]. Complete Tbx1 inactivation in pharyngeal endoderm is sufficient to cause a phenotype identical to the Tbx1 null mutants [219]. Part of the cardiovascular defect in Tbx1−/− mice reflects failed cardiac neural crest migration into the pharyngeal arches and heart where the crest produces signals such as RA that pattern heart structures [220]. Tbx1 in the otic vesicle is required for inner ear development, and also is essential for face and limb myoblast differentiation [213, 221223]. FGF signaling apparently influences Tbx1 function for pharyngeal arch derivatives [224]. Tbx1 heterozygosity is partially responsible for cranial nerve dysmorphology in the LgDel model of 22q11DS (Fig. 3d–g) [74]. Tbx1 heterozygotes have a significantly higher frequency of fusions and anastomoses between the glossopharyngeal (IX) and vagus (X) cranial nerves than WT embryos (Fig. 3e, f) [74]. This frequency is equivalent to that in LgDel embryos (Fig. 3d); however, Tbx1 mutants do not display the RA-sensitive LgDel trigeminal nerve (V) phenotype [74]. TBX1 heterozygous mono-allelic mutations have been associated with conotruncal anomaly face syndrome, thymic hypotrophy, parathyroid dysfunction, and deafness in a small sample of individuals without broader 22q11.2 deletion, providing a further suggestion of a key role in individuals with broader 22q11.2 deletion [225]. Finally, heterozygous Tbx1 mutation, like Gnb1l (an A to B protein trafficking gene; see above), reduces PPI in mice, indicating possible involvement in behavioral disorders [64]. This phenotypic equivalence, however, raises additional questions regarding the specificity of the PPI assay as well as a singular role for Tbx1 (or Gnb1l) in complex phenotypes, either in 22q11DS, or a wide range of animal models.
GSC2 Goosecoid Homeobox 2 (a.k.a. Goosecoid-like, GSCL; A to B) encodes a homeodomain-containing protein [226]. It is related to the homeobox gene, Goosecoid, which is required for craniofacial and rib development in mice [227]. Mouse Gsc2 is expressed in the neural tube and pharyngeal arches during neural crest migration and differentiation [228]. Nevertheless, homozygous Gsc2 mutant mice are viable with no obvious developmental anomalies [229]. As mentioned above, Gsc2 expression in the mature nervous system is restricted to the midbrain interpeduncular nucleus (IPN), implicated in dopaminergic modulation, sleep, and rapid eye movements. Despite this restricted expression, IPN-related phenotypes have not been reported in Gsc2−/− mice, except for loss of expression of an additional 22q11.2 A to B gene, the miRNA biogenesis factor Dgcr14 (see above) [190].
MED15 Mediator Complex Subunit 15 (a.k.a. ARC105; B to D) encodes a subunit of the mediator complex, a cofactor for RNA Polymerase II-dependent transcription. MED15 acts within the pre-initiation complex, which consists of MED15, POLII, TFIIA, TFIIB, TFIID, TFIIE, TFIIF, and TFIIH, and is considered a global regulator of gene expression [230, 231]. Mediator complex genes encode up to 30 subunits in some eukaryotes [231], and have limited homology between species. The MED15 subunit was initially identified based upon homology with Gal11, an essential transcriptional regulator of galactose metabolism in yeast [232], in which it may also function as a regulator of lipid homeostasis [233]. The C. elegans MED15 homolog, mdt-15 is required for fatty acid metabolism via transcription of genes that modulate desaturation of stearic to oleic acid [234]. Mdt-15 deficient worms move abnormally, are sterile, are more sensitive to stress, have disrupted ER homeostasis, and shortened life spans [234236]. In Drosophila, med15 is required for the transcription of decapentaplegic (dpp, homolog of vertebrate bone morphogenetic proteins (BMPs)) target genes [237]. The Xenopus MED15 homolog, arc105, similarly regulates transforming growth factor beta (TGFβ)/Activin/Nodal/Smad2/3 target gene expression [238]. MED15 function during mammalian development, however, remains elusive—there are no reported mouse mutants, nor are there any clinically described MED15 mutations, independent of heterozygous 22q11.2 A to D deletion, in humans.
KLHL22 Kelch Like Family Member 22 (B to D) encodes a member of the Bric-a-brac-Tramtrack-Broad complex (BTB)-Kelch transcription factor family that is conserved from Drosophila to humans [239, 240]. The BTB-Kelch proteins contain a BTB/POZ domain, a BACK domain, and five to seven Kelch motifs [239]. The BTB domains facilitate protein binding and have multiple cellular roles including recruitment of E3 ubiquitin ligase complex [241243]. Kelch domains form a β-propeller structure that, at least in some cases, interacts with actin and intermediate filaments and is involved in cytoskeleton organization [239, 244]. The BACK domain is a conserved motif found in the majority of proteins that contain both BTB and Kelch domains. Although no function has been reported for the BACK domain, it is likely to be of functional significance because mutations in this motif have been linked to human axonal neuropathy [245].
LZTR1 Leucine-Zipper-Like Transcription Regulator 1 (B to D) also encodes a member of the BTB-Kelch superfamily [244]. LZTR1 was initially described as a transcriptional regulator based on a weak homology to members of the basic leucine zipper-like family. However, further studies showed that LZTR1 localizes exclusively to the Golgi network where it helps stabilize the Golgi complex [246]. LZTR1 mutations are related to a rare genetic disorder called Schwannomatosis, characterized by multiple intracranial, spinal, and peripheral tumors (schwannomas) [247, 248]. Furthermore, biallelic pathogenic variants in LZTR1 have been linked to Noonan syndrome, a disorder characterized by unusual facial features, short stature, cardiovascular anomalies, bleeding, skeletal malformations, and developmental delays [249]. The parallels of Noonan/LZTR1 loss of function phenotypes with those in 22q11DS suggest it is a viable candidate for modulating core 22q11.2 anomalies in individuals with 3 Mb A to D deletions.
ZNF74 Zinc Finger Protein 74 (B to D) belongs to a large subfamily of C2-H2 (Cys2-His2) zinc finger proteins encoding Kruppel-associated box (KRAB) transcriptional repressor motifs [250]. In addition to DNA binding activity, a subset of the C2-H2 proteins bind RNAs and proteins [251, 252]. Family member TFIIIA binds to RNAs and proteins to regulate 5S rRNA transcription, storage, and transport from the nucleus to the cytoplasm [253, 254]. Similarly, ZNF74 encodes a nuclear matrix-attached protein with RNA binding properties [255]. ZNF74 interacts with a hyper-phosphorylated form of the largest subunit of RNA polymerase II and may regulate its activity during pre-mRNA processing [256]. There are no mouse or human genetic or functional analyses of this gene.
HIC2 Hypermethylated in Cancer 2 (a.k.a. ZBTB30 and ZNF907; B to D) is a BTB-zinc finger transcription factor required for normal heart development [257]. Homozygous Hic2 loss-of-function is early embryonic lethal and heterozygous mutants have ventricular septal defects and die at birth [258]. These anomalies implicate HIC2 as a potential modulator of 22q11DS phenotypes, particularly those in the heart, in typically deleted (A to D) individuals.
THAP7 Thanatos-Associated Protein 7 (B to D) encodes a member of a large family of THAP, an N-terminus 89-amino acid motif/C2-CH signature (Cys-X2–4-Cys-X35–53-Cys-X2-His) transcription factors [259]. THAP transcriptional regulators, found only in animal genomes [259, 260], are multifunctional transcriptional regulators. C. elegans THAP protein (HIM-17) is critical for chromosome segregation during meiosis [260]. The C2-CH signature zinc finger domain in THAP7 is similar to the site-specific DNA binding domain of Drosophila P-element transposase [259]. Human THAP7 functions as a transcriptional repressor by recruiting co-repressor NcoR and histone deacetylase HDAC3 to chromatin [261, 262]. Despite these intriguing functions, little is known about potential THAP7 contributions, based upon human mutations or animal models, to 22q11DS phenotypes.

Perspective: transcription factors

There are eight transcription factors located between LCRA and LCRD on hChr22q11.2: two in the A to B region, and six in the B to D region. Of these eight, TBX1 (A to B), HIC2 (B to D), and LZTR1 (B to D) when mutated in mice or humans influence heart and craniofacial phenotypes that have similarities to those in 22q11DS. Of all transcription factors, TBX1 has received the most attention as a “candidate” or even a single explanatory gene for 22q11DS phenotypes. TBX1 makes an essential contribution to 22q11DS cardiac, craniofacial, and otic phenotypes. Whether these contributions distinguish it formally as haploinsufficient for subsets of 22q11DS heart and face phenotypes versus a modulator of additional A to B or B to D gene interactions remains unresolved [73, 74]. Evidence for TBX1 modulation of brain development, particularly phenotypes crucial for 22q11DS behavioral deficits [211, 263] also is unresolved, although it is likely more limited than its contributions to cardiac or craniofacial differentiation [26]. In mouse, Tbx1 regulates distinct aspects of morphogenesis of cranial nerves IX and X (glossopharyngeal and vagal) that participate in respiratory control, feeding, and swallowing, suggesting that TBX1 may contribute to abnormalities in swallowing behavior observed in many individuals with 22q11DS [74, 264]. GSC2 (A to B) is not a likely contributor to 22q11DS pathology based upon available evidence. The role for dosage-dependent modulation of “core” (A to B-related) 22q11DS phenotypes of the remaining five B to D transcription factors could be significant, based on their apparent far-reaching effects on development and function. Nevertheless, almost no data, as yet, address possible interactions between modifying B to D transcription factors critical A to B genes.

Transmembrane receptors/transporters

RTN4R Reticulon 4 Receptor (a.k.a. Nogo receptor, Nogo-66 receptor, or NGR1; A to B). Of all the A to B genes, RTN4R is an intriguing candidate for nervous system phenotypes, and perhaps the most controversial. RTN4R, a neuronal glycosylphosphatidylinositol (GPI)-linked receptor, is expressed mainly in cerebral cortical neurons, hippocampal neurons, cerebellar Purkinje cells, and pontine neurons [265]. RTN4R binds to myelin-derived inhibitory proteins, MAG, OMgp, and Nogo isoforms and may limit neurite growth and axon regeneration after CNS injury in adult mammals [266269]. Consistent with these observations, in vivo studies report improved axon growth and locomotor recovery in spinal cord injured Rtn4r−/− mice or in injured rats treated with anti-Nogo-A antibodies [268, 270]. Adult rat motor cortical neurons can functionally reorganize after injury and treatment with anti-Nogo antibody, accompanied by increased movement of the lesion-impaired forelimb [271]. In the hippocampus, Rtnr4 restricts formation of both excitatory and inhibitory synapses, influences dendrite spine morphology and limits activity-dependent synaptic strength [272275]. In the visual cortex, Rtnr4 consolidates neural circuitry established during the visual critical period, and in other forebrain regions, it is required for consolidation of long-term memory [276278]. These results, however, have been challenged. Indeed, they have not been fully replicated and a great deal of evidence suggests that the Nogo/Rtn4r pathway alone makes at best a modest contribution to mammalian central nervous system regeneration and repair [279281]. In addition to its adult CNS expression, Rtn4r is expressed during brain development [282]. Nogo-A signaling via Rtn4r is implicated in ES cell self-renewal and differentiation [283285]. It may influence neural precursor migration in the developing and adult cerebral cortex [282, 286]. RTN4R also has been implicated as a susceptibility gene in psychosis through disruption of the brain microstructure [287, 288]. Several recent in vitro studies detect Rtn4r in immune cells, suggesting regulatory functions in immune cell adhesion and migration [289291]. The lack of clear data defining the function of Nogo-A/Rtn4r signaling makes its contribution to 22q11DS phenotypes uncertain.
P2RX6 Purinergic 2X6 Receptor (B to D) encodes a member of the P2X purinergic receptor family, which are ligand-gated ion channels activated by extracellular ATP, a neurotransmitter that causes fast excitatory postsynaptic potentials in neurons and smooth muscles [292295]. P2X receptors are trimers of P2X subunit proteins (P2X1–7) that associate as homomers or heteromers. The P2X6 subunit only assembles with P2X2 or P2X4 to form an active receptor complex [296, 297]. The P2rx6 gene was first isolated from a cDNA library from rat superior cervical ganglion cells [298], which are derived from the neural crest [299]. P2rx6 is expressed in the brain, localizes to glutamatergic synapses, increases during post-natal neurogenesis [300302], and modulates neuronal differentiation from neural progenitors in vitro [303]. Despite cellular evidence for P2rx6 function throughout the murine nervous system, there is no evidence for the effect of P2RX6 deficiency or null mutation either on mouse or human brain development.
SLC7A4 Solute Carrier Family 7 Member 4 (a.k.a. CAT4; B to D) is related by sequence to the cationic amino acid transporters (CAT) family [304]. In spite of its localization in the plasma membrane, SLC7A4 does not have transport activity when overexpressed in human cells or in Xenopus oocytes [305].

Perspective: transmembrane receptors/transporters

The one transmembrane receptor in the A to B region, RTN4R or NogoR, has potential nervous system functions that may contribute to 22q11DS phenotypes. Nevertheless, mouse heterozygous or homozygous mutants do not have reported phenotypes that parallel any of those associated with individuals with 22q11DS or mice with the orthologous A to B deletion [268, 280, 306]. Despite the still uncertain function of RTNR4 in neuronal plasticity or regeneration, it may interact with other A to B or B to D genes to modify 22q11DS phenotypes, particularly in the nervous system. Of the two B to D receptors, the P2RX6 is the most viable candidate for additional 22q11DS phenotypic variability.

Mitochondrial genes

PRODH Proline Dehydrogenase (a.k.a. Proline Oxidase; A to B) encodes a mitochondrial gene that catalyzes the first step of proline to glutamate conversion (Fig. 4a) [307, 308]. In the brain, Prodh is selectively expressed in the hypothalamus, amygdala, piriform cortex, hippocampus, and cerebral cortex (Figs. 1c and 4b, c) [58, 309311]. PRODH may be key for synaptic transmission. Proline may act as a neurotransmitter; however, the lack of a specific receptor in the brain suggests it may function instead as a neuromodulator [312, 313]. Indeed, in the mouse brain, proline may modulate glutamatergic synaptic transmission by directly activating glutamate receptors or potentiating transmission [314318]. In hyperprolinemic mice, memory deficits accompany elevated proline levels [319321]. Prodh−/− mice have sensorimotor gating defects and decreased hypothalamic glutamate and GABA [322]. PRODH loss of function mutations results in hyperprolinemia type 1, whose clinical symptoms largely depend on how severely proline function is reduced [307, 323]; while a minor increase in proline is mostly considered benign, high accumulation of proline is associated with severe phenotypes [323]. SCZ, intellectual disability, severe speech disorders, and seizures are associated with reduced PRODH activity due to polymorphisms or biallelic mutations independent of 22q11.2 deletion [323326]. A genome-wide association study, however, has challenged PRODH’s role as a risk factor for SCZ [327].
MRPL40 Mitochondrial Ribosomal Protein L40 (a.k.a. NLVCF; A to B) localizes to mitochondria and encodes the large subunit of the mitochondrial ribosome (Fig. 4a) [328]. Mrpl40 is expressed in the first and second pharyngeal arches in the mouse [329]. In the post-natal brain, Mrpl40 is enhanced in olfactory bulb mitral cells, cerebellar Purkinje cells, layer 5/6 cerebral cortex projection neurons, and the cortical subplate (Fig. 4b) [310], a transient early generated neuronal population between layer 5/6 and the cerebral ventricle that is necessary for initial thalamo-cortical and cortico-thalamic circuit formation [330, 331]. In the adult, Mrpl40 is enhanced in the same projection neuron classes (Fig. 4c) [310]. Heterozygous Mrpl40 mutants have synaptic plasticity and working memory deficits, altered mitochondrial permeability, increased presynaptic cytosolic Ca++, and enhanced short-term potentiation [332].
TANGO2 Transport and Golgi Organization 2 Homolog (a.k.a. T10; A to B) encodes a member of the transport and Golgi organization family that plays roles in ER secretory protein loading [333]. In Drosophila, tango2 loss-of-function leads to Golgi/ER fusion [333]. In contrast, mouse Tango2 has a mitochondrial localization motif and localizes to mitochondria, but not Golgi or peroxisomes (Fig. 4a) [310]. In the early postnatal mouse brain, Tango2 is restricted to olfactory bulb mitral cells (OB projection neurons), layer 5/6 cortical projection neurons, and the cortical subplate [310], and this restricted expression is maintained in the adult, except for the transient subplate (Fig. 4c). Mouse Tango2 mutants have not been reported. However, TANGO2 biallelic truncation mutations have been linked to a metabolic disorder, infancy-onset episodic metabolic crisis [334, 335], characterized by muscle weakness, recurrent rhabdomyolysis, cardiac arrhythmia, encephalopathy, progressive neurodegeneration, and cognitive impairment. In fibroblasts from an individual with a TANGO2 mutation, ER stress increases, Golgi volume and density decline, and mitochondrial fatty acid β-oxidation is aberrant [335]. Apparently, TANGO2 merits further assessment in humans and model systems as a contributor to the 22q11DS phenotypic spectrum.
ZDHHC8 Zinc Finger DHHC-Type Containing 8 (A to B) encodes a putative member of a family of transmembrane palmitoyl-transferases that share a conserved cysteine-rich catalytic domain (DHHC) [336]. It may modify multiple substrates including cell adhesion molecules, ion channel components, signaling, scaffold, and membrane-associated proteins [336]. Zdhhc8 has a mitochondrial signal sequence and localizes to mitochondria (Fig. 4a) [310]. In developing and adult mouse brain, Zdhhhc8 is enriched in the olfactory bulb, hippocampus, cerebellum, thalamic relay nuclei, and cerebral cortex (Figs. 1c, d and 4b, c) [58, 310, 311], and the protein is restricted to synapses in all of these regions. Zdhhc8 null mice are viable; however, hippocampal and cortical dendrite and axon growth as well as arborization, spine density, and glutamatergic synapse frequency declines [337, 338]. Some of these deficits can be rescued by enzymatically active ZDHHC8 [338]. Human ZDHHC8 palmitoylates PSD95, an adaptor involved in excitatory synapse development and plasticity [337, 339342] and PICK1, whose palmitoylation is necessary for cerebellar long-term synaptic depression [343]. ZDHHC8-dependent palmitoylation also is required for trafficking and stability of the D2 dopamine receptor protein to the plasma membrane [344]. ZDHHC8 is suggested to increase the risk of SCZ [345], although this claim was challenged in a recent study [327].
SLC25A1 Solute Carrier Family 25 Member 1 (a.k.a. CTP; A to B) encodes a highly conserved inner mitochondrial membrane transporter that mediates the movement of citrate/isocitrate in exchange for cytosolic malate [346, 347]. It modulates mitochondrial homeostasis, fatty acid and sterol biosynthesis, glycolysis, chromosome maintenance, and cytokine-dependent inflammatory responses [346, 348350]. In zebrafish, slc25a1 knockdown results in neuromuscular junction anomalies, axon outgrowth defects, and synaptic abnormality [351]. In humans, biallelic SLC25A1 mutations are associated with combined D-2- and L-2-hydroxyglutaric aciduria, a rare neurometabolic disorder characterized by hypotonia, epileptic encephalopathy, respiratory insufficiency, developmental arrest, severe neurodevelopmental dysfunction, lack of psychomotor development, and early death [352, 353]. Corpus callosum agenesis, optic nerve hypoplasia, and facial dysmorphism accompany these core clinical signs [354, 355]. SLC25A1 hemizygous mutation in combination with 22q11.2 deletion has been reported in an individual with severe brain, heart, and face phenotypes along with combined D-2- and L-2-hydroxyglutaric aciduria [356].
TXNRD2 Thioredoxin Reductase 2 (a.k.a. TR3; A to B) encodes a pyridine nucleotide-disulfide oxidoreductase family seleno-cysteine enzyme [357]. Txnrd2 mainly localizes to mitochondria; however, various N-terminal splicing variants may direct it to other cellular compartments [358]. Txnrd2 has two redox-active catalytic domains responsible for mitochondrial scavenging of reactive oxygen species essential for cell survival and mitochondria-mediated apoptosis [359361]. During mouse development, Txnrd2 is expressed in the heart and its constitutive loss-of-function leads to anemia, heart ventricular thinning, and fetal death [362]. Heart-specific Txnrd2 inactivation also elicits heart chamber dilation, swollen, structurally altered mitochondria, and death shortly after birth [362], reminiscent of human dilated developmental cardiomyopathy [363]. Heart-specific Txnrd2 inactivation in adult mice increases vulnerability to myocardial ischemia/reperfusion injury due to mitochondrial degeneration and contractile dysfunction [364, 365]. Even though oxidative stress is linked with neurodegeneration, nervous system-specific ablation of Txnrd2 does not cause apparent gross abnormalities in any region of the mouse brain [366]. In contrast to other studies that support the critical role of selenium and selenoproteins in immune responses [367, 368], T cell- and B cell-specific Txnrd2 disruption failed to phenocopy key defects in fetal hematopoiesis [369]. Txnrd2 loss-of-function phenotypes are similar to clinical features of Keshan Disease, caused by severe selenium deficiency [370].

Perspective: mitochondrial genes

22q11.2 mitochondrial genes are found only in the A to B region. These six genes: PRODH, MRPL40, TANGO2, ZDHHC8, SLC25A1, and TXNRD2 are implicated in cardiac, facial, and neural development, as well as brain function, and cognitive skills. The absence of B to D mitochondrial genes suggests 22q11DS bioenergetic/metabolic dysfunction may reflect primarily dosage change of the six A to B mitochondrial genes. Other B to D genes might modulate the functions of the six A to B mitochondrial genes, albeit indirectly. Nevertheless, single gene phenotypes in animal models and rare human homozygous loss-of-function mutations are all closely related to core 22q11DS phenotypes. Thus, a circumstantial case can be made for A to B mitochondrial gene contributions; however, this case needs to be strengthened by additional experimental and genetic evidence.

Hemostatic factors

GP1BB Glycoprotein Ib Platelet Beta Subunit (a.k.a. CD42C; A to B) encodes a subunit of the GP1b-V-IX complex, a hemostatic and thrombotic protein receptor primarily on platelet surfaces [371]. GP1BB mutations are associated with Bernard-Soulier syndrome [372], a rare disorder characterized by large platelets, low platelet count, and prolonged bleeding time [371]. Accordingly, there have been no assessments of Gp1bb mutant phenotypes in animal models.
SERPIND1 Serpin Family D Member 1 (a.k.a. Heparin Cofactor II; B to D), a serpin inflammation and coagulation superfamily modulator [373, 374], is a serine protease inhibitor that rapidly inactivates thrombin, a coagulation-related protease, and promotes angiogenesis and vascular remodeling after injury [375, 376]. Polymorphisms on the SERPIND1 promoter region may influence its expression [377]; however, no additional analyses confirm this finding.

Perspective: hemostatic factors

Hemostatic disruption has not been proposed as a frequent 22q11DS phenotype. Nevertheless, these anomalies are more common than previously thought [378380]. The disruption is mostly linked to GP1BB; however, there is a recent report that challenges this correlation [381]. It also possible that SERPIND1 located in B to D region modifies the function of GP1BB the in A to D deleted individuals, especially if regulatory polymorphisms further vary expression levels of this B to D gene.

Additional genes with unique functions

DGCR 6 DiGeorge Critical Region 6 (A to B) is encoded by two highly homologous ORFs (DGCR6 and DGCR6L) [382]. DGCR6 has homology to Gdl, a gonadal development related protein in Drosophila, and the human laminin gamma-1 subunit, LAMC1, which is essential for basal lamina assembly [383]. Although distinct functions for the two ORFs encoding DGCR6 variants have not yet been fully defined, DGCR6 may influence neural crest migration and pharyngeal arch development [58, 384, 385]. In chick embryos, DGCR6 suppression results in cardiovascular anomalies [384]. In these embryos, attenuation of DGCR6 stimulates TBX1 and UFD1L but decreases heart and pharyngeal arch HIRA expression (all A to B genes), suggesting it is a modifier of critical region phenotypes [384]. DGCR6 also has been implicated in 22q11DS conotruncal heart defects, either directly or through TBX1 activity [386]. Dysregulation of DGCR6 and DGCR6L is suggested to be associated with neuropsychological findings in 22q11DS children [387].
UFD1L Ubiquitin Fusion-Degradation Protein-1 Like (a.k.a. UFD1; A to B) is a component of the Ufd1L-Npl4-p97 multiprotein complex that recognizes and presents several polyubiquitin-tagged proteins to the proteasome for degradation [388, 389]. IP3 receptors, downstream components of PI4KA pathway (see above), are included among Ufd1L-Npl4-p97 targets [390]. The Ufd1-Npl4-p97 complex is required for clearance of damaged mitochondria [391, 392]. UFD1L also participates in ER-associated degradation, spindle disassembly, DNA-damage response, cell-cycle control, telomerase length regulation, and ribosome-associated degradation [393397]. UFD1L functional attenuation in chick cardiac neural crest increases conotruncal septation defects [398]. Ufd1l is expressed in pharyngeal arches, palatal precursors, and developing ears, heart, and brain (Fig. 1c, d) [188, 399]. Ufd1l−/− embryos die before organogenesis [398]. In human, UFD1L expression is reduced in bicuspid aortic valve, a common cardiac defect, further supporting a role in proper heart formation [400]. Drosophila ufd1 is implicated in neuronal function and maintenance [401]. Its role in mouse and human brain function is less clear. In the LgDel brain, Ufd1L transcripts decline by approximately 50%, but Ufd1L protein is at WT levels [311]. UFD1L polymorphisms are associated with increased corpus callosum volumes and have been linked to cognitive deficits independent of 22q11.2 deletion [402, 403].
COMT Catechol-O-Methyltransferase (A to B) is one of several enzymes that degrade catecholamines, including dopamine, epinephrine, and norepinephrine [404]. COMT membrane-bound and soluble isoforms are predominately expressed in brain and peripheral tissues, respectively [404, 405]. Comt is expressed in several brain regions (Fig. 1c, d). Mutant mice lacking Comt show impaired emotional reactivity and cognition as well as increased aggressive behaviors [406, 407]. COMT polymorphisms are linked independently to SCZ, bipolar disorder, obsessive-compulsive disorder, anorexia, and ADHD [20, 408410]—many are also 22q11DS behavioral phenotypes. This correlation suggests a potential role for COMT in regulating dopamine levels, which are disrupted in most of these disorders [411]. COMT association to SCZ, however, was challenged in a recent genome-wide association study [327].
TSSK2 Testis Specific Serine Kinase 2 (a.k.a. STK22b; A to B) encodes a serine/threonine kinase involved in spermatogenesis [412]. In mouse, the Stk22b orthologue (STK22a is a pseudogene) is not expressed at 22q11.2 phenotypic sites (heart, face, limbs, thymus, brain), nor detected in the maturing or adult brain [58]. Recently, one patient with 22q11DS has been reported to have azoospermia, which could reflect TSSK2 deficiency [413].
TRMT2a tRNA Methyltransferase 2 Homolog A (a.k.a. HTF9C; A to B) is a tRNA methyltransferase with homology to a yeast scTrm2 DNA repair enzyme [414]. TRMT2a is one of 34 human tRNA methyltransferases that target approximately 600 tRNA variants encoded by the human genome [415] to modulate tRNA stability, translational efficiency, and protein production. TRMT2A may contribute to 22q11DS phenotypes; however, genomic diversity of tRNA methyltransferases and their tRNA targets raises the possibility of functional redundancy and compensation. Trmt2a is expressed in the heart, pharyngeal arches, and brain (Fig. 1c) [58]. TRMT2a and its murine orthologue share a bidirectional promoter with Ranbp1, an established 22q11.2 contributor gene (see above); however, Trmt2a is expressed at WT levels in Ranbp1−/− mice [170]. Promoter polymorphisms in the TRMT2A/RANBP1 locus, however, could yield additional dosage-dependent changes in 22q11DS.

Perspective: additional genes with unique functions

All “sui generis” 22q11.2 genes are in the A to B region, implicating each as a potential contributor to key phenotypes. Of the four, TSSK2 seems least likely to contribute to neural, cardiovascular, or craniofacial development. Similarly, UFD1L, if translationally dosage compensated in humans as in mice, is also less likely. DGCR6 merits further consideration following additional mechanistic studies. COMT remains a prime suspect based upon its contributions to aminergic neural signaling and potential association with behavioral disorders.

Conclusion

Single suspects and accomplices from the 22q11.2 gene line-up

Over the last decade, data addressing developmental roles of several 22q11.2 genes have increased dramatically based on extrapolation of full loss-of-function phenotypes caused by deletion of single 22q11.2 orthologues in mouse and other model species. Nevertheless, the relationships between altered dosage of 22q11.2 genes—rather than homozygous loss of function—and the many and variable phenotypes in individuals with 22q11DS remain unclear. The spectrum of speculations regarding which gene “explains” 22q11.2 phenotypes is as broad as the 22q11DS phenotypic spectrum itself. Proposed explanations range from single gene haploinsufficiency accounting for all essential 22q11DS phenotypes, to more nuanced contiguous gene or gene-gene interactions that lead to subsets of clinically significant features. Several “genes of interest” have been identified; however, the potential contribution of several others—both confirmed coding loci as well as unannotated open reading frames with uncertain protein coding function as well as microRNAs—have not been thoroughly considered. We revisited the “line-up” of deleted 22q11.2 genes—focusing on those that encode identified proteins detected in at least one tissue in humans or model organisms—in the 3 Mb, typically deleted, A to D region. Our assessment of primary cellular functions and mutant dysfunction of all known protein coding 22q11.2 genes allows us to address two unsolved questions essential for understanding 22q11DS pathogenic mechanisms: (1) which phenotypes reflect true haploinsufficiency for A to B or A to D genes? (2) What causes phenotypic variability in individuals with distinct or even identical deletions? This 22q11.2 gene “line-up,” reflecting insight from human and model system genetic analysis as well as parallel cell biological and molecular assessments establishes a genetic interaction matrix (Fig. 5) that generates multiple outcomes with differing severity (Fig. 6). Understanding the detailed nature of this matrix, we suggest, will yield better therapeutic options for clinical challenges faced by individuals with 22q11DS. Additional investigation is necessary to complete the suspect list and decipher interactions that modify or diversify key 22q11DS features. Future studies targeting gene “cabals” in animal models should elucidate additional gene-gene interactions within the phenotypic matrix defined by A to B and B to D deletions (Figs. 5 and 6). Nevertheless, this matrix alone cannot reliably predict combinations of dosage-dependent phenotypes due to 22q11.2 deletions. Indeed, these predictions might be generated by combining the 22q11.2 interaction matrix with data on individual polymorphisms beyond 22q11.2 genes as well as fetal and post-natal environmental exposures [73, 264].

Candidate genes

“Which gene?” remains a central question for CNV-associated developmental disorders. If 22q11DS diagnostic assessment and therapies are to be targeted to pathogenic processes and delivered at appropriate times to maximize benefits in 22q11DS and other CNV-associated syndromes, it is imperative to identify culpable genes, characterize their obligate functions, and relate those data to disease-causing mechanisms and individual variability. Based upon evidence gathered by multiple investigators, it is unlikely that haploinsufficiency of a single 22q11.2 gene accounts for the full range of 22q11DS-related phenotypes. Nevertheless, our “line-up” indicates new promising leads on mechanistic interactions between suspects that disrupt development or homeostasis in 22q11DS. At least three possibilities—and one red herring—emerge from our functional genomic synthesis of minimally and typically deleted 22q11.2 genes.

The red herring

A single gene explains core 22q11DS phenotypes (Fig. 6a). Beyond doubt, several A to B as well as B to D genes, due to loss-of-function mutations or diminished dosage, contribute substantially, yet variably, to 22q11DS-related phenotypes. Nevertheless, the conclusion that any single gene “acts alone” to cause even one, let alone the full spectrum of 22q11DS phenotypes, without being influenced by diminished dosage of others, is no longer tenable based on judicious assessment of the evidence summarized here. What, then, more viable leads emerge from our thorough investigation of individual 22q11.2 genes and the primary cellular and developmental function of the proteins they encode?

First

22q11DS is a classic “contiguous gene syndrome” (Fig. 6b) in which diminished dosage of single A to B genes independently leads to each key 22q11.2 phenotype: one for the brain, one for the heart, one for the face, and one for the thymus. In this scenario, B to D genes may modulate each A to B gene’s singular influence, but do not account for the core phenotypic change. Differing phenotypic frequencies in single gene heterozygous mutants versus broader deletion (see Fig. 3), or stochastic interactions between A to B and B to D genes and signals like sonic hedgehog (Shh), RA, or FGF [73, 74, 217, 416] suggest this is unlikely.

Second

Gene subsets form multigenic “syndicates” to disrupt critical developmental mechanisms or homeostatic function at specific phenotypic sites (Fig. 6c). For example, diminished dosage of Crkl (B to D), combined with that of Tbx1 (A to B) and a few other A to B genes may be responsible for many of the core cardiovascular anomalies as well as their variability [417]. Other distinct groups of 22q11 genes might similarly impact the brain, face, or thymus. There is considerable evidence for these sorts of selective, phenotype-specific interactions, at least in the heart, and most of this evidence has been reviewed here.

Third

A broader “interactome” is impacted by dosage-related changes in which proteins of similar cellular function encoded by multiple A to B genes, modulated by B to D genes, act in concert to disrupt essential cellular mechanisms at several phenotypic sites, rather than to elicit one single phenotype. These interactions may be due to sequential disruption of cell biological mechanisms leading to a threshold of developmental or homeostatic dysfunction at multiple phenotypic sites, or more direct, but locally distinct, interactions among functionally related 22q11.2 proteins in different organs and tissues. Such fundamental 22q11.2 dosage-dependent interactions may then be further modified by an individual’s genomic background independent of 22q11.2 deletion (Fig. 6d). In this scenario, 22q11.2 genes with similar function, particularly mitochondrial, transcription factor, adhesion molecules, or signaling factors—some only in the A to B interval, others from B to D—disrupt cellular mechanisms common to development and/or maintenance of heart, face, immune, skeletal, and brain integrity.
Additional evidence supports this “interactome” hypothesis as a viable solution to the yet unsolved mystery of 22q11.2 dosage-dependent phenotypic change and variability. Within the 22q11.2 deleted region, ARVCF and RANBP1 (A to B) as well as CRKL and SNAP29 (B to D) are predicted to interact with cadherin 1 (CDH1). Such interactions, influenced by stoichiometric change in proteins encoded by 22q11.2 genes, and modulated by polymorphisms in CDH1 in this example, or additional loci for other subsets of functionally related 22q11.2 proteins, could generally alter adhesion-dependent morphogenesis at all of 22q11DS phenotypic sites. CRKL (B to D), implicated in TBX1-dependent phenotypic changes, also interacts with phosphatase and tensin homolog (PTEN), a lipid phosphatase in the PI3K-AKT signaling pathway, involved in cell growth, proliferation, survival, and migration [418]. In addition, CRKL as well as DGCR8 interact with proteins involved in mRNA maturation and degradation, further supporting convergence on distinct cellular mechanisms that could compromise multiple phenotypic sites as more plausible mechanism for 22q11DS pathogenesis. The global instability of transcriptional regulation and its stochastic effects on 22q11 genes, perhaps effectively lowering their expression levels below a “loss of function” threshold on a cell-by-cell basis might broadly compromise multiple steps of development and maintenance of the heart, thymus, limbs, face, and brain. Analysis of genomic, transcriptomic, and proteomic databases can generate additional hypotheses of this sort of cellular mechanistic convergence. These hypotheses can then be tested genetically in the most appropriate model system among the several described here, or analyzed in cell lines, or in induced pluripotent stem cells (iPSCs) from several individuals with 22q11.2 deletions to further assess variability. Critically, one would predict that experiments in which key modulators of the cell biological mechanisms upon which subsets of 22q11.2 genes converge (see Fig. 6) were manipulated genetically or pharmacologically would provide further confirmation of the third scenario.
Our gene dosage-based/convergence-upon-cellular-mechanism hypothesis for 22q11DS is generally consistent with recent genomic analyses in non-syndromic individuals diagnosed with two behavioral disorders that are also significantly associated with 22q11DS: ASD and SCZ. There may be similar convergence on essential cellular mechanisms in ASD: 65 risk genes have been identified as chromatin regulators or establishment and maintenance of synaptic stability [21]. Multiple genes that encode cell adhesion molecules, tight junction proteins, and regulators of vesicle trafficking—all involved in critical processes for neural circuit development and neuronal signaling—have been implicated in SCZ susceptibility [22, 23]. Apparently, multigenic origins of complex brain disorders like ASD and SCZ may reflect a combination of single gene variants that independently are benign, but as a group, target common cellular mechanisms leading to dysfunction. Viewing these variants as independent actors, rather than interactors in a network, might result in incomplete explanations of pathology in clinically defined diseases like ASD, SCZ, or in multi-gene CNV syndromes like 22q11DS. New efforts to evaluate concerted actions of multiple genes on focal mechanisms for brain development and function are needed to advance our understanding of neurodevelopmental disorders generally. Our newly assembled gene 22q11.2 “line-up,” from LCR A to LCR D, allows us to view “suspect” genes anew and formulate new investigative strategies for 22q11DS, perhaps providing a template for understanding a wider range of multigenic or CNV-associated syndromes.

Acknowledgements

We thank members of the Moody and LaMantia laboratories for helpful discussions during the preparation of this manuscript.

Funding

Work in the authors’ laboratories on 22q11DS has been supported by NIH HD042182 (ASL), HD083157 (A-S.L. and SAM), and Simons Foundation SFARI 306796 (ASL) and SFARI 342005(TM).

Availability of data and materials

There is no original data generated in the authors’ laboratories presented in this review article. The supporting data is comprised entirely of references to published literature freely available in publicly accessible databases, particularly those maintained by the National Library of Medicine (PubMed, OMIM) as well as several genomic databases for which URLs are provided in the text of the manuscript.

Authors’ information

Zahra Motahari, Ph.D. Post-doctoral Associated, Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037 motahariz@gwu.​edu
Sally A. Moody, Ph.D. Professor and Chair, Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037.
samoody@gwu.​edu
Thomas M. Maynard Ph.D., Associate Research Professor, Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037.
maynard@gwu.​edu
Anthony-Samuel LaMantia, Ph.D. Lieberman Professor of Neuroscience, Director, GW Institute for Neuroscience, Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington DC, 20037.
lamantia@gwu.​edu
There are no data from human subject studies requiring IRB review or patient consent in this article. There are no original data collected for or presented in the paper.
All four of the co-authors (Motahari, Moody, Maynard, LaMantia) read and approved the manuscript for submission for publication.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat McDonald-McGinn DM, Sullivan KE. Chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/ velocardiofacial syndrome). Medicine (Baltimore). 2011;90(1):1–18.CrossRef McDonald-McGinn DM, Sullivan KE. Chromosome 22q11.2 deletion syndrome (DiGeorge syndrome/ velocardiofacial syndrome). Medicine (Baltimore). 2011;90(1):1–18.CrossRef
2.
Zurück zum Zitat Burn J, Takao A, Wilson D, Cross I, Momma K, Wadey R, Scambler P, Goodship J. Conotruncal anomaly face syndrome is associated with a deletion within chromosome 22q11. J Med Genet. 1993;30(10):822–4.CrossRefPubMedPubMedCentral Burn J, Takao A, Wilson D, Cross I, Momma K, Wadey R, Scambler P, Goodship J. Conotruncal anomaly face syndrome is associated with a deletion within chromosome 22q11. J Med Genet. 1993;30(10):822–4.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Fernandez A, Meechan D, Baker JL, Karpinski BA, LaMantia AS, Maynard TM. 22q11 deletion syndrome: copy number variations and development. In: Principles of Developmental Genetics. 2nd ed; 2015. p. 677–96.CrossRef Fernandez A, Meechan D, Baker JL, Karpinski BA, LaMantia AS, Maynard TM. 22q11 deletion syndrome: copy number variations and development. In: Principles of Developmental Genetics. 2nd ed; 2015. p. 677–96.CrossRef
4.
Zurück zum Zitat Robin NH, Shprintzen RJ. Defining the clinical spectrum of deletion 22q11.2. J Pediatr. 2005;147(1):90–6.CrossRefPubMed Robin NH, Shprintzen RJ. Defining the clinical spectrum of deletion 22q11.2. J Pediatr. 2005;147(1):90–6.CrossRefPubMed
5.
Zurück zum Zitat Schneider M, Debbane M, Bassett AS, Chow EW, Fung WL, van den Bree M, et al. Psychiatric disorders from childhood to adulthood in 22q11.2 deletion syndrome: results from the international consortium on brain and behavior in 22q11.2 deletion syndrome. Am J Psychiatry. 2014;171(6):627–39.CrossRefPubMedPubMedCentral Schneider M, Debbane M, Bassett AS, Chow EW, Fung WL, van den Bree M, et al. Psychiatric disorders from childhood to adulthood in 22q11.2 deletion syndrome: results from the international consortium on brain and behavior in 22q11.2 deletion syndrome. Am J Psychiatry. 2014;171(6):627–39.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Shaikh TH, Kurahashi H, Emanuel BS. Evolutionarily conserved low copy repeats (LCRs) in 22q11 mediate deletions, duplications, translocations, and genomic instability: an update and literature review. Genet Med. 2001;3(1):6–13.CrossRefPubMed Shaikh TH, Kurahashi H, Emanuel BS. Evolutionarily conserved low copy repeats (LCRs) in 22q11 mediate deletions, duplications, translocations, and genomic instability: an update and literature review. Genet Med. 2001;3(1):6–13.CrossRefPubMed
7.
Zurück zum Zitat Griffiths AJF. Modern genetic analysis : integrating genes and genomes, vol. xix. 2nd ed. New York: W.H. Freeman and Co; 2002. p. 736. Griffiths AJF. Modern genetic analysis : integrating genes and genomes, vol. xix. 2nd ed. New York: W.H. Freeman and Co; 2002. p. 736.
9.
Zurück zum Zitat Carlson C, Sirotkin H, Pandita R, Goldberg R, McKie J, Wadey R, et al. Molecular definition of 22q11 deletions in 151 velo-cardio-facial syndrome patients. Am J Hum Genet. 1997;61(3):620–9.CrossRefPubMedPubMedCentral Carlson C, Sirotkin H, Pandita R, Goldberg R, McKie J, Wadey R, et al. Molecular definition of 22q11 deletions in 151 velo-cardio-facial syndrome patients. Am J Hum Genet. 1997;61(3):620–9.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Michaelovsky E, Frisch A, Carmel M, Patya M, Zarchi O, Green G, et al. Genotype-phenotype correlation in 22q11.2 deletion syndrome. BMC Med Genet. 2012;13:122.CrossRefPubMedPubMedCentral Michaelovsky E, Frisch A, Carmel M, Patya M, Zarchi O, Green G, et al. Genotype-phenotype correlation in 22q11.2 deletion syndrome. BMC Med Genet. 2012;13:122.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Nogueira SI, Hacker AM, Bellucco FT, Christofolini DM, Kulikowski LD, Cernach MC, et al. Atypical 22q11.2 deletion in a patient with DGS/VCFS spectrum. Eur J Med Genet. 2008;51(3):226–30.CrossRefPubMedPubMedCentral Nogueira SI, Hacker AM, Bellucco FT, Christofolini DM, Kulikowski LD, Cernach MC, et al. Atypical 22q11.2 deletion in a patient with DGS/VCFS spectrum. Eur J Med Genet. 2008;51(3):226–30.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Burnside RD. 22q11.21 deletion syndromes: a review of proximal, central, and distal deletions and their associated features. Cytogenet Genome Res. 2015;146(2):89–99.CrossRefPubMed Burnside RD. 22q11.21 deletion syndromes: a review of proximal, central, and distal deletions and their associated features. Cytogenet Genome Res. 2015;146(2):89–99.CrossRefPubMed
13.
Zurück zum Zitat Racedo SE, McDonald-McGinn DM, Chung JH, Goldmuntz E, Zackai E, Emanuel BS, et al. Mouse and human CRKL is dosage sensitive for cardiac outflow tract formation. Am J Hum Genet. 2015;96(2):235–44.CrossRefPubMedPubMedCentral Racedo SE, McDonald-McGinn DM, Chung JH, Goldmuntz E, Zackai E, Emanuel BS, et al. Mouse and human CRKL is dosage sensitive for cardiac outflow tract formation. Am J Hum Genet. 2015;96(2):235–44.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Michaelovsky E, Frisch A, Carmel M, Patya M, Zarchi O, Green T, et al. Genotype-phenotype correlation in 22q11.2 deletion syndrome. BMC Med Genet. 2012;13:122.CrossRefPubMedPubMedCentral Michaelovsky E, Frisch A, Carmel M, Patya M, Zarchi O, Green T, et al. Genotype-phenotype correlation in 22q11.2 deletion syndrome. BMC Med Genet. 2012;13:122.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Vergaelen E, Swillen A, Van Esch H, Claes S, Van Goethem G, Devriendt K. 3 generation pedigree with paternal transmission of the 22q11.2 deletion syndrome: Intrafamilial phenotypic variability. Eur J Med Genet. 2015;58(4):244–8.CrossRefPubMed Vergaelen E, Swillen A, Van Esch H, Claes S, Van Goethem G, Devriendt K. 3 generation pedigree with paternal transmission of the 22q11.2 deletion syndrome: Intrafamilial phenotypic variability. Eur J Med Genet. 2015;58(4):244–8.CrossRefPubMed
16.
Zurück zum Zitat Guo T, McDonald-McGinn D, Blonska A, Shanske A, Bassett AS, Chow E, et al. Genotype and cardiovascular phenotype correlations with TBX1 in 1,022 velo-cardio-facial/DiGeorge/22q11.2 deletion syndrome patients. Hum Mutat. 2011;32(11):1278–89.CrossRefPubMedPubMedCentral Guo T, McDonald-McGinn D, Blonska A, Shanske A, Bassett AS, Chow E, et al. Genotype and cardiovascular phenotype correlations with TBX1 in 1,022 velo-cardio-facial/DiGeorge/22q11.2 deletion syndrome patients. Hum Mutat. 2011;32(11):1278–89.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Clements CC, Wenger TL, Zoltowski AR, Bertollo JR, Miller JS, de Marchena AB, et al. Critical region within 22q11.2 linked to higher rate of autism spectrum disorder. Mol Autism. 2017;8:58.CrossRefPubMedPubMedCentral Clements CC, Wenger TL, Zoltowski AR, Bertollo JR, Miller JS, de Marchena AB, et al. Critical region within 22q11.2 linked to higher rate of autism spectrum disorder. Mol Autism. 2017;8:58.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Hiroi N, Takahashi T, Hishimoto A, Izumi T, Boku S, Hiramoto T. Copy number variation at 22q11.2: from rare variants to common mechanisms of developmental neuropsychiatric disorders. Mol Psychiatry. 2013;18(11):1153–65.CrossRefPubMed Hiroi N, Takahashi T, Hishimoto A, Izumi T, Boku S, Hiramoto T. Copy number variation at 22q11.2: from rare variants to common mechanisms of developmental neuropsychiatric disorders. Mol Psychiatry. 2013;18(11):1153–65.CrossRefPubMed
19.
Zurück zum Zitat Jungerius BJ, Hoogendoorn ML, Bakker SC, Van't Slot R, Bardoel AF, Ophoff RA, et al. An association screen of myelin-related genes implicates the chromosome 22q11 PIK4CA gene in schizophrenia. Mol Psychiatry. 2008;13(11):1060–8.CrossRefPubMed Jungerius BJ, Hoogendoorn ML, Bakker SC, Van't Slot R, Bardoel AF, Ophoff RA, et al. An association screen of myelin-related genes implicates the chromosome 22q11 PIK4CA gene in schizophrenia. Mol Psychiatry. 2008;13(11):1060–8.CrossRefPubMed
20.
Zurück zum Zitat Prasad SE, Howley S, Murphy KC. Candidate genes and the behavioral phenotype in 22q11.2 deletion syndrome. Dev Disabil Res Rev. 2008;14(1):26–34.CrossRefPubMed Prasad SE, Howley S, Murphy KC. Candidate genes and the behavioral phenotype in 22q11.2 deletion syndrome. Dev Disabil Res Rev. 2008;14(1):26–34.CrossRefPubMed
21.
Zurück zum Zitat Sanders SJ, He X, Willsey AJ, Ercan-Sencicek AG, Samocha KE, Cicek AE, et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron. 2015;87(6):1215–33.CrossRefPubMedPubMedCentral Sanders SJ, He X, Willsey AJ, Ercan-Sencicek AG, Samocha KE, Cicek AE, et al. Insights into autism spectrum disorder genomic architecture and biology from 71 risk loci. Neuron. 2015;87(6):1215–33.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Purcell SM, Moran JL, Fromer M, Ruderfer D, Solovieff N, Roussos P, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506(7487):185–90.CrossRefPubMedPubMedCentral Purcell SM, Moran JL, Fromer M, Ruderfer D, Solovieff N, Roussos P, et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature. 2014;506(7487):185–90.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat O'Dushlaine C, Kenny E, Heron E, Donohoe G, Gill M, Morris D, et al. Molecular pathways involved in neuronal cell adhesion and membrane scaffolding contribute to schizophrenia and bipolar disorder susceptibility. Mol Psychiatry. 2011;16(3):286–92.CrossRefPubMed O'Dushlaine C, Kenny E, Heron E, Donohoe G, Gill M, Morris D, et al. Molecular pathways involved in neuronal cell adhesion and membrane scaffolding contribute to schizophrenia and bipolar disorder susceptibility. Mol Psychiatry. 2011;16(3):286–92.CrossRefPubMed
24.
Zurück zum Zitat Sifrim A, Hitz MP, Wilsdon A, Breckpot J, Turki SH, Thienpont B, et al. Distinct genetic architectures for syndromic and nonsyndromic congenital heart defects identified by exome sequencing. Nat Genet. 2016;48(9):1060–5.CrossRefPubMedPubMedCentral Sifrim A, Hitz MP, Wilsdon A, Breckpot J, Turki SH, Thienpont B, et al. Distinct genetic architectures for syndromic and nonsyndromic congenital heart defects identified by exome sequencing. Nat Genet. 2016;48(9):1060–5.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Guna A, Butcher NJ, Bassett AS. Comparative mapping of the 22q11.2 deletion region and the potential of simple model organisms. J Neurodev Disord. 2015;7(1):18.CrossRefPubMedPubMedCentral Guna A, Butcher NJ, Bassett AS. Comparative mapping of the 22q11.2 deletion region and the potential of simple model organisms. J Neurodev Disord. 2015;7(1):18.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Meechan DW, Maynard TM, Tucker ES, Fernandez A, Karpinski BA, Rothblat LA, et al. Modeling a model: mouse genetics, 22q11.2 deletion syndrome, and disorders of cortical circuit development. Prog Neurobiol. 2015;130:1–28.CrossRefPubMedPubMedCentral Meechan DW, Maynard TM, Tucker ES, Fernandez A, Karpinski BA, Rothblat LA, et al. Modeling a model: mouse genetics, 22q11.2 deletion syndrome, and disorders of cortical circuit development. Prog Neurobiol. 2015;130:1–28.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Ray-Gallet D, Quivy J, Scamps C, Martini EM, Lipinski M, Almouzni G. HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol Cell. 2002;9(5):1091–100.CrossRefPubMed Ray-Gallet D, Quivy J, Scamps C, Martini EM, Lipinski M, Almouzni G. HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol Cell. 2002;9(5):1091–100.CrossRefPubMed
28.
Zurück zum Zitat Hall C, Nelson DM, Ye X, Baker K, DeCaprio JA, Seeholzer S, et al. HIRA, the human homologue of yeast Hir1p and Hir2p, is a novel cyclin-cdk2 substrate whose expression blocks S-phase progression. Mol Cell Biol. 2001;21(5):1854–65.CrossRefPubMedPubMedCentral Hall C, Nelson DM, Ye X, Baker K, DeCaprio JA, Seeholzer S, et al. HIRA, the human homologue of yeast Hir1p and Hir2p, is a novel cyclin-cdk2 substrate whose expression blocks S-phase progression. Mol Cell Biol. 2001;21(5):1854–65.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Spector MS, Raff A, DeSilva H, Lee K, Osley MA. Hir1p and Hir2p function as transcriptional corepressors to regulate histone gene transcription in the Saccharomyces cerevisiae cell cycle. Mol Cell Biol. 1997;17(2):545–52.CrossRefPubMedPubMedCentral Spector MS, Raff A, DeSilva H, Lee K, Osley MA. Hir1p and Hir2p function as transcriptional corepressors to regulate histone gene transcription in the Saccharomyces cerevisiae cell cycle. Mol Cell Biol. 1997;17(2):545–52.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Anderson HE, Wardle J, Korkut SV, Murton HE, Lopez-Maury L, Bahler J, et al. The fission yeast HIRA histone chaperone is required for promoter silencing and the suppression of cryptic antisense transcripts. Mol Cell Biol. 2009;29(18):5158–67.CrossRefPubMedPubMedCentral Anderson HE, Wardle J, Korkut SV, Murton HE, Lopez-Maury L, Bahler J, et al. The fission yeast HIRA histone chaperone is required for promoter silencing and the suppression of cryptic antisense transcripts. Mol Cell Biol. 2009;29(18):5158–67.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Rai TS, Cole JJ, Nelson DM, Dikovskaya D, Faller WJ, Vizioli MG, et al. HIRA orchestrates a dynamic chromatin landscape in senescence and is required for suppression of neoplasia. Genes Dev. 2014;28(24):2712–25.CrossRefPubMedPubMedCentral Rai TS, Cole JJ, Nelson DM, Dikovskaya D, Faller WJ, Vizioli MG, et al. HIRA orchestrates a dynamic chromatin landscape in senescence and is required for suppression of neoplasia. Genes Dev. 2014;28(24):2712–25.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat van der Heijden GW, Derijck AA, Posfai E, Giele M, Pelczar P, Ramos L, et al. Chromosome-wide nucleosome replacement and H3.3 incorporation during mammalian meiotic sex chromosome inactivation. Nat Genet. 2007;39(2):251–8.CrossRefPubMed van der Heijden GW, Derijck AA, Posfai E, Giele M, Pelczar P, Ramos L, et al. Chromosome-wide nucleosome replacement and H3.3 incorporation during mammalian meiotic sex chromosome inactivation. Nat Genet. 2007;39(2):251–8.CrossRefPubMed
33.
Zurück zum Zitat Zhang R, Poustovoitov MV, Ye X, Santos HA, Chen W, Daganzo SM, et al. Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev Cell. 2005;8(1):19–30.CrossRefPubMed Zhang R, Poustovoitov MV, Ye X, Santos HA, Chen W, Daganzo SM, et al. Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev Cell. 2005;8(1):19–30.CrossRefPubMed
34.
Zurück zum Zitat Ahmad A, Kikuchi H, Takami Y, Nakayama T. Different roles of N-terminal and C-terminal halves of HIRA in transcription regulation of cell cycle-related genes that contribute to control of vertebrate cell growth. J Biol Chem. 2005;280(37):32090–100.CrossRefPubMed Ahmad A, Kikuchi H, Takami Y, Nakayama T. Different roles of N-terminal and C-terminal halves of HIRA in transcription regulation of cell cycle-related genes that contribute to control of vertebrate cell growth. J Biol Chem. 2005;280(37):32090–100.CrossRefPubMed
35.
Zurück zum Zitat Ahmad K, Henikoff S. The histone variant H3.3 marks active chromatin by replication-independent nucleosome assembly. Mol Cell. 2002;9(6):1191–200.CrossRefPubMed Ahmad K, Henikoff S. The histone variant H3.3 marks active chromatin by replication-independent nucleosome assembly. Mol Cell. 2002;9(6):1191–200.CrossRefPubMed
36.
Zurück zum Zitat Dutta D, Ray S, Home P, Saha B, Wang S, Sheibani N, et al. Regulation of angiogenesis by histone chaperone HIRA-mediated incorporation of lysine 56-acetylated histone H3.3 at chromatin domains of endothelial genes. J Biol Chem. 2010;285(53):41567–77.CrossRefPubMedPubMedCentral Dutta D, Ray S, Home P, Saha B, Wang S, Sheibani N, et al. Regulation of angiogenesis by histone chaperone HIRA-mediated incorporation of lysine 56-acetylated histone H3.3 at chromatin domains of endothelial genes. J Biol Chem. 2010;285(53):41567–77.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Pchelintsev NA, McBryan T, Rai TS, van Tuyn J, Ray-Gallet D, Almouzni G, et al. Placing the HIRA histone chaperone complex in the chromatin landscape. Cell Rep. 2013;3(4):1012–9.CrossRefPubMedPubMedCentral Pchelintsev NA, McBryan T, Rai TS, van Tuyn J, Ray-Gallet D, Almouzni G, et al. Placing the HIRA histone chaperone complex in the chromatin landscape. Cell Rep. 2013;3(4):1012–9.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Adam S, Polo SE, Almouzni G. Transcription recovery after DNA damage requires chromatin priming by the H3.3 histone chaperone HIRA. Cell. 2013;155(1):94–106.CrossRefPubMed Adam S, Polo SE, Almouzni G. Transcription recovery after DNA damage requires chromatin priming by the H3.3 histone chaperone HIRA. Cell. 2013;155(1):94–106.CrossRefPubMed
39.
Zurück zum Zitat Mandemaker IK, Vermeulen W, Marteijn JA. Gearing up chromatin: a role for chromatin remodeling during the transcriptional restart upon DNA damage. Nucleus. 2014;5(3):203–10.CrossRefPubMedPubMedCentral Mandemaker IK, Vermeulen W, Marteijn JA. Gearing up chromatin: a role for chromatin remodeling during the transcriptional restart upon DNA damage. Nucleus. 2014;5(3):203–10.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Yang JH, Song TY, Jo C, Park J, Lee HY, Song I, et al. Differential regulation of the histone chaperone HIRA during muscle cell differentiation by a phosphorylation switch. Exp Mol Med. 2016;48:e252.CrossRefPubMedPubMedCentral Yang JH, Song TY, Jo C, Park J, Lee HY, Song I, et al. Differential regulation of the histone chaperone HIRA during muscle cell differentiation by a phosphorylation switch. Exp Mol Med. 2016;48:e252.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Roberts C, Sutherland HF, Farmer H, Kimber W, Halford S, Carey A, et al. Targeted mutagenesis of the Hira gene results in gastrulation defects and patterning abnormalities of mesoendodermal derivatives prior to early embryonic lethality. Mol Cell Biol. 2002;22(7):2318–28.CrossRefPubMedPubMedCentral Roberts C, Sutherland HF, Farmer H, Kimber W, Halford S, Carey A, et al. Targeted mutagenesis of the Hira gene results in gastrulation defects and patterning abnormalities of mesoendodermal derivatives prior to early embryonic lethality. Mol Cell Biol. 2002;22(7):2318–28.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Roberts C, Daw SC, Halford S, Scambler PJ. Cloning and developmental expression analysis of chick Hira (Chira), a candidate gene for DiGeorge syndrome. Hum Mol Genet. 1997;6(2):237–45.CrossRefPubMed Roberts C, Daw SC, Halford S, Scambler PJ. Cloning and developmental expression analysis of chick Hira (Chira), a candidate gene for DiGeorge syndrome. Hum Mol Genet. 1997;6(2):237–45.CrossRefPubMed
43.
Zurück zum Zitat Magnaghi P, Roberts C, Lorain S, Lipinski M, Scambler PJ. HIRA, a mammalian homologue of Saccharomyces cerevisiae transcriptional co-repressors, interacts with Pax3. Nat Genet. 1998;20(1):74–7.CrossRefPubMed Magnaghi P, Roberts C, Lorain S, Lipinski M, Scambler PJ. HIRA, a mammalian homologue of Saccharomyces cerevisiae transcriptional co-repressors, interacts with Pax3. Nat Genet. 1998;20(1):74–7.CrossRefPubMed
44.
Zurück zum Zitat Conway SJ, Henderson DJ, Copp AJ. Pax3 is required for cardiac neural crest migration in the mouse: evidence from the splotch (Sp2H) mutant. Development. 1997;124(2):505–14.PubMed Conway SJ, Henderson DJ, Copp AJ. Pax3 is required for cardiac neural crest migration in the mouse: evidence from the splotch (Sp2H) mutant. Development. 1997;124(2):505–14.PubMed
45.
Zurück zum Zitat Szenker E, Lacoste N, Almouzni G. A developmental requirement for HIRA-dependent H3.3 deposition revealed at gastrulation in Xenopus. Cell Rep. 2012;1(6):730–40.CrossRefPubMed Szenker E, Lacoste N, Almouzni G. A developmental requirement for HIRA-dependent H3.3 deposition revealed at gastrulation in Xenopus. Cell Rep. 2012;1(6):730–40.CrossRefPubMed
46.
Zurück zum Zitat Wilming LG, Snoeren CA, van Rijswijk A, Grosveld F, Meijers C. The murine homologue of HIRA, a DiGeorge syndrome candidate gene, is expressed in embryonic structures affected in human CATCH22 patients. Hum Mol Genet. 1997;6(2):247–58.CrossRefPubMed Wilming LG, Snoeren CA, van Rijswijk A, Grosveld F, Meijers C. The murine homologue of HIRA, a DiGeorge syndrome candidate gene, is expressed in embryonic structures affected in human CATCH22 patients. Hum Mol Genet. 1997;6(2):247–58.CrossRefPubMed
47.
Zurück zum Zitat Farrell MJ, Stadt H, Wallis KT, Scambler P, Hixon RL, Wolfe R, Leatherbury L, Kirby ML. HIRA, a DiGeorge syndrome candidate gene, is required for cardiac outflow tract septation. Circ Res. 1999;84(2):127–35.CrossRefPubMed Farrell MJ, Stadt H, Wallis KT, Scambler P, Hixon RL, Wolfe R, Leatherbury L, Kirby ML. HIRA, a DiGeorge syndrome candidate gene, is required for cardiac outflow tract septation. Circ Res. 1999;84(2):127–35.CrossRefPubMed
48.
Zurück zum Zitat Valenzuela N, Fan Q, Fa'ak F, Soibam B, Nagandla H, Liu Y, et al. Cardiomyocyte-specific conditional knockout of the histone chaperone HIRA in mice results in hypertrophy, sarcolemmal damage and focal replacement fibrosis. Dis Model Mech. 2016;9(3):335–45.CrossRefPubMedPubMedCentral Valenzuela N, Fan Q, Fa'ak F, Soibam B, Nagandla H, Liu Y, et al. Cardiomyocyte-specific conditional knockout of the histone chaperone HIRA in mice results in hypertrophy, sarcolemmal damage and focal replacement fibrosis. Dis Model Mech. 2016;9(3):335–45.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Broderick R, Nasheuer HP. Regulation of Cdc45 in the cell cycle and after DNA damage. Biochem Soc Trans. 2009;37(Pt 4):926–30.CrossRefPubMed Broderick R, Nasheuer HP. Regulation of Cdc45 in the cell cycle and after DNA damage. Biochem Soc Trans. 2009;37(Pt 4):926–30.CrossRefPubMed
50.
Zurück zum Zitat Machida YJ, Hamlin JL, Dutta A. Right place, right time, and only once: replication initiation in metazoans. Cell. 2005;123(1):13–24.CrossRefPubMed Machida YJ, Hamlin JL, Dutta A. Right place, right time, and only once: replication initiation in metazoans. Cell. 2005;123(1):13–24.CrossRefPubMed
51.
Zurück zum Zitat Kang YH, Galal WC, Farina A, Tappin I, Hurwitz J. Properties of the human Cdc45/Mcm2-7/GINS helicase complex and its action with DNA polymerase epsilon in rolling circle DNA synthesis. Proc Natl Acad Sci U S A. 2012;109(16):6042–7.CrossRefPubMedPubMedCentral Kang YH, Galal WC, Farina A, Tappin I, Hurwitz J. Properties of the human Cdc45/Mcm2-7/GINS helicase complex and its action with DNA polymerase epsilon in rolling circle DNA synthesis. Proc Natl Acad Sci U S A. 2012;109(16):6042–7.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Pacek M, Tutter AV, Kubota Y, Takisawa H, Walter JC. Localization of MCM2-7, Cdc45, and GINS to the site of DNA unwinding during eukaryotic DNA replication. Mol Cell. 2006;21(4):581–7.CrossRefPubMed Pacek M, Tutter AV, Kubota Y, Takisawa H, Walter JC. Localization of MCM2-7, Cdc45, and GINS to the site of DNA unwinding during eukaryotic DNA replication. Mol Cell. 2006;21(4):581–7.CrossRefPubMed
54.
Zurück zum Zitat Zou L, Mitchell J, Stillman B. CDC45, a novel yeast gene that functions with the origin recognition complex and mcm proteins in initiation of DNA replication. Mol Cell Biol. 1997;17(2):553–63.CrossRefPubMedPubMedCentral Zou L, Mitchell J, Stillman B. CDC45, a novel yeast gene that functions with the origin recognition complex and mcm proteins in initiation of DNA replication. Mol Cell Biol. 1997;17(2):553–63.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Kohler C, Koalick D, Fabricius A, Parplys AC, Borgmann K, Pospiech H, et al. Cdc45 is limiting for replication initiation in humans. Cell Cycle. 2016;15(7):974–85.CrossRefPubMedPubMedCentral Kohler C, Koalick D, Fabricius A, Parplys AC, Borgmann K, Pospiech H, et al. Cdc45 is limiting for replication initiation in humans. Cell Cycle. 2016;15(7):974–85.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Edwards MC, Tutter AV, Cvetic C, Gilbert CH, Prokhorova TA, Walter JC. MCM2-7 complexes bind chromatin in a distributed pattern surrounding the origin recognition complex in Xenopus egg extracts. J Biol Chem. 2002;277(36):33049–57.CrossRefPubMed Edwards MC, Tutter AV, Cvetic C, Gilbert CH, Prokhorova TA, Walter JC. MCM2-7 complexes bind chromatin in a distributed pattern surrounding the origin recognition complex in Xenopus egg extracts. J Biol Chem. 2002;277(36):33049–57.CrossRefPubMed
58.
Zurück zum Zitat Maynard TM, Haskell GT, Peters AZ, Sikich L, Lieberman JA, LaMantia AS. A comprehensive analysis of 22q11 gene expression in the developing and adult brain. Proc Natl Acad Sci U S A. 2003;100(24):14433–8.CrossRefPubMedPubMedCentral Maynard TM, Haskell GT, Peters AZ, Sikich L, Lieberman JA, LaMantia AS. A comprehensive analysis of 22q11 gene expression in the developing and adult brain. Proc Natl Acad Sci U S A. 2003;100(24):14433–8.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Shaikh TH, Gottlieb S, Sellinger B, Chen F, Roe BA, Oakey RJ, et al. Characterization of CDC45L: a gene inthe 22q11.2 deletion region expressed during murine and human development. Mamm Genome. 1999;10(3):322–6.CrossRefPubMed Shaikh TH, Gottlieb S, Sellinger B, Chen F, Roe BA, Oakey RJ, et al. Characterization of CDC45L: a gene inthe 22q11.2 deletion region expressed during murine and human development. Mamm Genome. 1999;10(3):322–6.CrossRefPubMed
60.
Zurück zum Zitat Meechan DW, Tucker ES, Maynard TM, LaMantia AS. Diminished dosage of 22q11 genes disrupts neurogenesis and cortical development in a mouse model of 22q11 deletion/DiGeorge syndrome. Proc Natl Acad Sci U S A. 2009;106(38):16434–45.CrossRefPubMedPubMedCentral Meechan DW, Tucker ES, Maynard TM, LaMantia AS. Diminished dosage of 22q11 genes disrupts neurogenesis and cortical development in a mouse model of 22q11 deletion/DiGeorge syndrome. Proc Natl Acad Sci U S A. 2009;106(38):16434–45.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Yoshida K, Kuo F, George EL, Sharpe AH, Dutta A. Requirement of CDC45 for postimplantation mouse development. Mol Cell Biol. 2001;21(14):4598–603.CrossRefPubMedPubMedCentral Yoshida K, Kuo F, George EL, Sharpe AH, Dutta A. Requirement of CDC45 for postimplantation mouse development. Mol Cell Biol. 2001;21(14):4598–603.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Fenwick AL, Kliszczak M, Cooper F, Murray J, Sanchez-Pulido L, Twigg SR, et al. Mutations in CDC45, encoding an essential component of the pre-initiation complex, cause Meier-Gorlin syndrome and craniosynostosis. Am J Hum Genet. 2016;99(1):125–38.CrossRefPubMedPubMedCentral Fenwick AL, Kliszczak M, Cooper F, Murray J, Sanchez-Pulido L, Twigg SR, et al. Mutations in CDC45, encoding an essential component of the pre-initiation complex, cause Meier-Gorlin syndrome and craniosynostosis. Am J Hum Genet. 2016;99(1):125–38.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Gong L, Liu M, Jen J, Yeh ET. GNB1L, a gene deleted in the critical region for DiGeorge syndrome on 22q11 encodes a G-protein beta-subunit-like polypeptide. Biochim Biophys Acta. 2000;1494(1–2):185–8.CrossRefPubMed Gong L, Liu M, Jen J, Yeh ET. GNB1L, a gene deleted in the critical region for DiGeorge syndrome on 22q11 encodes a G-protein beta-subunit-like polypeptide. Biochim Biophys Acta. 2000;1494(1–2):185–8.CrossRefPubMed
64.
Zurück zum Zitat Paylor R, Glaser B, Mupo A, Ataliotis P, Spencer C, Sobotka A, et al. Tbx1 haploinsufficiency is linked to behavioral disorders in mice and humans: implications for 22q11 deletion syndrome. Proc Natl Acad Sci U S A. 2006;103(20):7729–34.CrossRefPubMedPubMedCentral Paylor R, Glaser B, Mupo A, Ataliotis P, Spencer C, Sobotka A, et al. Tbx1 haploinsufficiency is linked to behavioral disorders in mice and humans: implications for 22q11 deletion syndrome. Proc Natl Acad Sci U S A. 2006;103(20):7729–34.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Feller S. Crk family adaptors -signalling complex formation and biological roles. Oncogene. 2001;20:6348–71.CrossRefPubMed Feller S. Crk family adaptors -signalling complex formation and biological roles. Oncogene. 2001;20:6348–71.CrossRefPubMed
66.
Zurück zum Zitat Birge RB, Kalodimos C, Inagaki F, Tanaka S. Crk and CrkL adaptor proteins: networks for physiological and pathological signaling. Cell Commun Signal. 2009;7:13.CrossRefPubMedPubMedCentral Birge RB, Kalodimos C, Inagaki F, Tanaka S. Crk and CrkL adaptor proteins: networks for physiological and pathological signaling. Cell Commun Signal. 2009;7:13.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Ballif BA, Arnaud L, Arthur WT, Guris D, Imamoto A, Cooper JA. Activation of a Dab1/CrkL/C3G/Rap1 pathway in Reelin-stimulated neurons. Curr Biol. 2004;14(7):606–10.CrossRefPubMed Ballif BA, Arnaud L, Arthur WT, Guris D, Imamoto A, Cooper JA. Activation of a Dab1/CrkL/C3G/Rap1 pathway in Reelin-stimulated neurons. Curr Biol. 2004;14(7):606–10.CrossRefPubMed
68.
Zurück zum Zitat Feng L, Cooper JA. Dual functions of Dab1 during brain development. Mol Cell Biol. 2009;29(2):324–32.CrossRefPubMed Feng L, Cooper JA. Dual functions of Dab1 during brain development. Mol Cell Biol. 2009;29(2):324–32.CrossRefPubMed
69.
Zurück zum Zitat Huang Y, Magdaleno S, Hopkins R, Slaughter C, Curran T, Keshvara L. Tyrosine phosphorylated disabled 1 recruits Crk family adapter proteins. Biochem Biophys Res Commun. 2004;318(1):204–12.CrossRefPubMed Huang Y, Magdaleno S, Hopkins R, Slaughter C, Curran T, Keshvara L. Tyrosine phosphorylated disabled 1 recruits Crk family adapter proteins. Biochem Biophys Res Commun. 2004;318(1):204–12.CrossRefPubMed
70.
Zurück zum Zitat Guris DL, Fantes J, Tara D, Druker BJ, Imamoto A. Mice lacking the homologue of the human 22q11.2 geneCRKL phenocopy neurocristopathies of DiGeorge syndrome. Nat Genet. 2001;27(3):293–8.CrossRefPubMed Guris DL, Fantes J, Tara D, Druker BJ, Imamoto A. Mice lacking the homologue of the human 22q11.2 geneCRKL phenocopy neurocristopathies of DiGeorge syndrome. Nat Genet. 2001;27(3):293–8.CrossRefPubMed
71.
Zurück zum Zitat Miller KA, Tan TY, Welfare MF, White SM, Stark Z, Savarirayan R, et al. A mouse splice-site mutant and individuals with atypical chromosome 22q11.2 deletions demonstrate the crucial role for crkl in craniofacial and pharyngeal development. Mol Syndromol. 2014;5(6):276–86.CrossRefPubMedPubMedCentral Miller KA, Tan TY, Welfare MF, White SM, Stark Z, Savarirayan R, et al. A mouse splice-site mutant and individuals with atypical chromosome 22q11.2 deletions demonstrate the crucial role for crkl in craniofacial and pharyngeal development. Mol Syndromol. 2014;5(6):276–86.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Guris DL, Duester G, Papaioannou VE, Imamoto A. Dose-dependent interaction of Tbx1 and Crkl and locally aberrant RA signaling in a model of del22q11 syndrome. Dev Cell. 2006;10(1):81–92.CrossRefPubMed Guris DL, Duester G, Papaioannou VE, Imamoto A. Dose-dependent interaction of Tbx1 and Crkl and locally aberrant RA signaling in a model of del22q11 syndrome. Dev Cell. 2006;10(1):81–92.CrossRefPubMed
73.
Zurück zum Zitat Maynard TM, Gopalakrishna D, Meechan DW, Paronett EM, Newbern JM, LaMantia AS. 22q11 gene dosage establishes an adaptive range for sonic hedgehog and retinoic acid signaling during early development. Hum Mol Genet. 2013;22(2):300–12.CrossRefPubMed Maynard TM, Gopalakrishna D, Meechan DW, Paronett EM, Newbern JM, LaMantia AS. 22q11 gene dosage establishes an adaptive range for sonic hedgehog and retinoic acid signaling during early development. Hum Mol Genet. 2013;22(2):300–12.CrossRefPubMed
74.
Zurück zum Zitat Karpinski BA, Maynard TM, Fralish MS, Nuwayhid S, Zohn IE, Moody SA, et al. Dysphagia and disrupted cranial nerve development in a mouse model of DiGeorge (22q11) deletion syndrome. Dis Model Mech. 2014;7(2):245–57.CrossRefPubMed Karpinski BA, Maynard TM, Fralish MS, Nuwayhid S, Zohn IE, Moody SA, et al. Dysphagia and disrupted cranial nerve development in a mouse model of DiGeorge (22q11) deletion syndrome. Dis Model Mech. 2014;7(2):245–57.CrossRefPubMed
75.
Zurück zum Zitat Moon AM, Guris DL, Seo JH, Li L, Hammond J, Talbot A, et al. Crkl deficiency disrupts Fgf8 signaling in a mouse model of 22q11 deletion syndromes. Dev Cell. 2006;10(1):71–80.CrossRefPubMedPubMedCentral Moon AM, Guris DL, Seo JH, Li L, Hammond J, Talbot A, et al. Crkl deficiency disrupts Fgf8 signaling in a mouse model of 22q11 deletion syndromes. Dev Cell. 2006;10(1):71–80.CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat Giacomelli M, Kumar R, Soresina A, Tamassia N, Lorenzini T, Moratto D, et al. Reduction of CRKL expression in patients with partial DiGeorge syndrome is associated with impairment of T-cell functions. J Allergy Clin Immunol. 2016;138(1):229–40 e3.CrossRefPubMed Giacomelli M, Kumar R, Soresina A, Tamassia N, Lorenzini T, Moratto D, et al. Reduction of CRKL expression in patients with partial DiGeorge syndrome is associated with impairment of T-cell functions. J Allergy Clin Immunol. 2016;138(1):229–40 e3.CrossRefPubMed
77.
Zurück zum Zitat Zheng P, Noroski LM, Hanson IC, Chen Y, Lee ME, Huang Y, et al. Molecular mechanisms of functional natural killer deficiency in patients with partial DiGeorge syndrome. J Allergy Clin Immunol. 2015;135(5):1293–302.CrossRefPubMedPubMedCentral Zheng P, Noroski LM, Hanson IC, Chen Y, Lee ME, Huang Y, et al. Molecular mechanisms of functional natural killer deficiency in patients with partial DiGeorge syndrome. J Allergy Clin Immunol. 2015;135(5):1293–302.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Huang Y, Clarke F, Karimi M, Roy NH, Williamson EK, Okumura M, et al. CRK proteins selectively regulate T cell migration into inflamed tissues. J Clin Invest. 2015;125(3):1019–32.CrossRefPubMedPubMedCentral Huang Y, Clarke F, Karimi M, Roy NH, Williamson EK, Okumura M, et al. CRK proteins selectively regulate T cell migration into inflamed tissues. J Clin Invest. 2015;125(3):1019–32.CrossRefPubMedPubMedCentral
79.
80.
Zurück zum Zitat Fish EN, Uddin S, Korkmaz M, Majchrzak B, Druker BJ, Platanias LC. Activation of a CrkL-stat5 signaling complex by type I interferons. J Biol Chem. 1999;274(2):571–3.CrossRefPubMed Fish EN, Uddin S, Korkmaz M, Majchrzak B, Druker BJ, Platanias LC. Activation of a CrkL-stat5 signaling complex by type I interferons. J Biol Chem. 1999;274(2):571–3.CrossRefPubMed
81.
Zurück zum Zitat Ishii J, Adachi H, Aoki J, Koizumi H, Tomita S, Suzuki T, et al. SREC-II, a new member of the scavenger receptor type F family, trans-interacts with SREC-I through its extracellular domain. J Biol Chem. 2002;277(42):39696–702.CrossRefPubMed Ishii J, Adachi H, Aoki J, Koizumi H, Tomita S, Suzuki T, et al. SREC-II, a new member of the scavenger receptor type F family, trans-interacts with SREC-I through its extracellular domain. J Biol Chem. 2002;277(42):39696–702.CrossRefPubMed
82.
Zurück zum Zitat Hwang M, Kalinin A, Morasso MI. The temporal and spatial expression of the novel ca++-binding proteins, scarf and Scarf2, during development and epidermal differentiation. Gene Expr Patterns. 2005;5(6):801–8.CrossRefPubMedPubMedCentral Hwang M, Kalinin A, Morasso MI. The temporal and spatial expression of the novel ca++-binding proteins, scarf and Scarf2, during development and epidermal differentiation. Gene Expr Patterns. 2005;5(6):801–8.CrossRefPubMedPubMedCentral
83.
Zurück zum Zitat Anastasio N, Ben-Omran T, Teebi A, Ha KC, Lalonde E, Ali R, et al. Mutations in SCARF2 are responsible for Van Den Ende-Gupta syndrome. Am J Hum Genet. 2010;87(4):553–9.CrossRefPubMedPubMedCentral Anastasio N, Ben-Omran T, Teebi A, Ha KC, Lalonde E, Ali R, et al. Mutations in SCARF2 are responsible for Van Den Ende-Gupta syndrome. Am J Hum Genet. 2010;87(4):553–9.CrossRefPubMedPubMedCentral
84.
Zurück zum Zitat Migliavacca MP, Sobreira NL, Antonialli GP, Oliveira MM, Melaragno MI, Casteels I, et al. Sclerocornea in a patient with van den Ende-Gupta syndrome homozygous for a SCARF2 microdeletion. Am J Med Genet A. 2014;164A(5):1170–4.CrossRefPubMed Migliavacca MP, Sobreira NL, Antonialli GP, Oliveira MM, Melaragno MI, Casteels I, et al. Sclerocornea in a patient with van den Ende-Gupta syndrome homozygous for a SCARF2 microdeletion. Am J Med Genet A. 2014;164A(5):1170–4.CrossRefPubMed
85.
Zurück zum Zitat Schweitzer DN, Lachman RS, Pressman BD, Graham JM Jr. Van den Ende-Gupta syndrome of blepharophimosis, arachnodactyly, and congenital contractures: clinical delineation and recurrence in brothers. Am J Med Genet A. 2003;118A(3):267–73.CrossRefPubMed Schweitzer DN, Lachman RS, Pressman BD, Graham JM Jr. Van den Ende-Gupta syndrome of blepharophimosis, arachnodactyly, and congenital contractures: clinical delineation and recurrence in brothers. Am J Med Genet A. 2003;118A(3):267–73.CrossRefPubMed
86.
Zurück zum Zitat Balla A, Balla T. Phosphatidylinositol 4-kinases: old enzymes with emerging functions. Trends Cell Biol. 2006;16(7):351–61.CrossRefPubMed Balla A, Balla T. Phosphatidylinositol 4-kinases: old enzymes with emerging functions. Trends Cell Biol. 2006;16(7):351–61.CrossRefPubMed
87.
Zurück zum Zitat Audhya A, Foti M, Emr SD. Distinct roles for the yeast phosphatidylinositol 4- kinases, Stt4p and Pik1p, in secretion, cell growth, and organelle membrane dynamics. Mol Biol Cell. 2000;11(8):2673–89.CrossRefPubMedPubMedCentral Audhya A, Foti M, Emr SD. Distinct roles for the yeast phosphatidylinositol 4- kinases, Stt4p and Pik1p, in secretion, cell growth, and organelle membrane dynamics. Mol Biol Cell. 2000;11(8):2673–89.CrossRefPubMedPubMedCentral
88.
Zurück zum Zitat Heilmeyer L, Vereb G, Vereb G, Kakuk A, Szivák I. Mammalian phosphatidylinositol 4-kinases. IUBMB Life. 2003;55(2):59–65.CrossRefPubMed Heilmeyer L, Vereb G, Vereb G, Kakuk A, Szivák I. Mammalian phosphatidylinositol 4-kinases. IUBMB Life. 2003;55(2):59–65.CrossRefPubMed
89.
Zurück zum Zitat Nakagawa T, Goto K, Kondo H. Cloning, expression, and localization of 230-kDa phosphatidylinositol 4- kinase. J Biol Chem. 1996;271(20):12088–94.CrossRefPubMed Nakagawa T, Goto K, Kondo H. Cloning, expression, and localization of 230-kDa phosphatidylinositol 4- kinase. J Biol Chem. 1996;271(20):12088–94.CrossRefPubMed
90.
Zurück zum Zitat Wong K. Meyers ddR, Cantley LC. Subcellular locations of phosphatidylinositol 4-kinase isoforms. J Biol Chem. 1997;272(20):13236–41.CrossRefPubMed Wong K. Meyers ddR, Cantley LC. Subcellular locations of phosphatidylinositol 4-kinase isoforms. J Biol Chem. 1997;272(20):13236–41.CrossRefPubMed
91.
92.
Zurück zum Zitat Nakatsu F, Baskin JM, Chung J, Tanner LB, Shui G, Lee SY, et al. PtdIns4P synthesis by PI4KIIIalpha at the plasma membrane and its impact on plasma membrane identity. J Cell Biol. 2012;199(6):1003–16.CrossRefPubMedPubMedCentral Nakatsu F, Baskin JM, Chung J, Tanner LB, Shui G, Lee SY, et al. PtdIns4P synthesis by PI4KIIIalpha at the plasma membrane and its impact on plasma membrane identity. J Cell Biol. 2012;199(6):1003–16.CrossRefPubMedPubMedCentral
93.
Zurück zum Zitat Ma H, Blake T, Chitnis A, Liu P, Balla T. Crucial role of phosphatidylinositol 4-kinase IIIalpha in development of zebrafish pectoral fin is linked to phosphoinositide 3-kinase and FGF signaling. J Cell Sci. 2009;122(Pt 23):4303–10.CrossRefPubMedPubMedCentral Ma H, Blake T, Chitnis A, Liu P, Balla T. Crucial role of phosphatidylinositol 4-kinase IIIalpha in development of zebrafish pectoral fin is linked to phosphoinositide 3-kinase and FGF signaling. J Cell Sci. 2009;122(Pt 23):4303–10.CrossRefPubMedPubMedCentral
94.
Zurück zum Zitat Wong K, Cantley LC. Cloning and characterization of a human phosphatidylinositol 4-kinase. J Biol Chem. 1994;269(46):28878–84.PubMed Wong K, Cantley LC. Cloning and characterization of a human phosphatidylinositol 4-kinase. J Biol Chem. 1994;269(46):28878–84.PubMed
95.
Zurück zum Zitat Zólyomi A, Zhao X, Downing GJ, Balla T. Localization of two distinct type III phosphatidylinositol 4-kinase enzyme mRNAs in the rat. Am J Physiol Cell Physiol. 2000;278(5):C914–20.CrossRefPubMed Zólyomi A, Zhao X, Downing GJ, Balla T. Localization of two distinct type III phosphatidylinositol 4-kinase enzyme mRNAs in the rat. Am J Physiol Cell Physiol. 2000;278(5):C914–20.CrossRefPubMed
96.
Zurück zum Zitat Bojjireddy N, Botyanszki J, Hammond G, Creech D, Peterson R, Kemp DC, et al. Pharmacological and genetic targeting of the PI4KA enzyme reveals its important role in maintaining plasma membrane.phosphatidylinositol 4-phosphate and phosphatidylinositol 4,5-bisphosphate levels. J Biol Chem. 2014;289(9):6120–32.CrossRefPubMedPubMedCentral Bojjireddy N, Botyanszki J, Hammond G, Creech D, Peterson R, Kemp DC, et al. Pharmacological and genetic targeting of the PI4KA enzyme reveals its important role in maintaining plasma membrane.phosphatidylinositol 4-phosphate and phosphatidylinositol 4,5-bisphosphate levels. J Biol Chem. 2014;289(9):6120–32.CrossRefPubMedPubMedCentral
97.
Zurück zum Zitat Vaillancourt FH, Brault M, Pilote L, Uyttersprot N, Gaillard ET, Stoltz JH, et al. Evaluation of phosphatidylinositol-4-kinase IIIalpha as a hepatitis C virus drug target. J Virol. 2012;86(21):11595–607.CrossRefPubMedPubMedCentral Vaillancourt FH, Brault M, Pilote L, Uyttersprot N, Gaillard ET, Stoltz JH, et al. Evaluation of phosphatidylinositol-4-kinase IIIalpha as a hepatitis C virus drug target. J Virol. 2012;86(21):11595–607.CrossRefPubMedPubMedCentral
98.
Zurück zum Zitat Pagnamenta AT, Howard MF, Wisniewski E, Popitsch N, Knight SJ, Keays DA, et al. Germline recessive mutations in PI4KA are associated with perisylvian polymicrogyria,cerebellar hypoplasia and arthrogryposis. Hum Mol Genet. 2015;24(13):3732–41.CrossRefPubMedPubMedCentral Pagnamenta AT, Howard MF, Wisniewski E, Popitsch N, Knight SJ, Keays DA, et al. Germline recessive mutations in PI4KA are associated with perisylvian polymicrogyria,cerebellar hypoplasia and arthrogryposis. Hum Mol Genet. 2015;24(13):3732–41.CrossRefPubMedPubMedCentral
99.
Zurück zum Zitat Vorstman JA, Chow EW, Ophoff RA, van Engeland H, Beemer FA, Kahn RS, et al. Association of the PIK4CA schizophrenia-susceptibility gene in adults with the 22q11.2 deletion syndrome. Am J Med Genet B Neuropsychiatr Genet. 2009;150B(3):430–3.CrossRefPubMedPubMedCentral Vorstman JA, Chow EW, Ophoff RA, van Engeland H, Beemer FA, Kahn RS, et al. Association of the PIK4CA schizophrenia-susceptibility gene in adults with the 22q11.2 deletion syndrome. Am J Med Genet B Neuropsychiatr Genet. 2009;150B(3):430–3.CrossRefPubMedPubMedCentral
100.
Zurück zum Zitat Carnahan RH, Rokas A, Gaucher EA, Reynolds AB. The molecular evolution of the p120-catenin subfamily and its functional associations. PLoS One. 2010;5(12):e15747.CrossRefPubMedPubMedCentral Carnahan RH, Rokas A, Gaucher EA, Reynolds AB. The molecular evolution of the p120-catenin subfamily and its functional associations. PLoS One. 2010;5(12):e15747.CrossRefPubMedPubMedCentral
102.
Zurück zum Zitat Kausalya PJ, Phua DC, Hunziker W. Association of ARVCF with zonula occludens (ZO)-1 and ZO-2: binding to PDZ-domain proteins and cell-cell adhesion regulate plasma membrane and nuclear localization of ARVCF. Mol Biol Cell. 2004;15(12):5503–15.CrossRefPubMedPubMedCentral Kausalya PJ, Phua DC, Hunziker W. Association of ARVCF with zonula occludens (ZO)-1 and ZO-2: binding to PDZ-domain proteins and cell-cell adhesion regulate plasma membrane and nuclear localization of ARVCF. Mol Biol Cell. 2004;15(12):5503–15.CrossRefPubMedPubMedCentral
103.
Zurück zum Zitat Laura RP, Witt AS, Held HA, Gerstner R, Deshayes K, Koehler MF, et al. The Erbin PDZ domain binds with high affinity and specificity to the carboxyl termini of delta-catenin and ARVCF. J Biol Chem. 2002;277(15):12906–14.CrossRefPubMed Laura RP, Witt AS, Held HA, Gerstner R, Deshayes K, Koehler MF, et al. The Erbin PDZ domain binds with high affinity and specificity to the carboxyl termini of delta-catenin and ARVCF. J Biol Chem. 2002;277(15):12906–14.CrossRefPubMed
104.
Zurück zum Zitat Sirotkin H, O'Donnell H, DasGupta R, Halford S, St Jore B, Puech A, et al. Identification of a new human catenin gene family member (ARVCF) from the region deleted in velo-cardio-facial syndrome. Genomics. 1997;41(1):75–83.CrossRefPubMed Sirotkin H, O'Donnell H, DasGupta R, Halford S, St Jore B, Puech A, et al. Identification of a new human catenin gene family member (ARVCF) from the region deleted in velo-cardio-facial syndrome. Genomics. 1997;41(1):75–83.CrossRefPubMed
106.
Zurück zum Zitat Kaufmann U, Zuppinger C, Waibler Z, Rudiger M, Urbich C, Martin B, et al. The armadillo repeat region targets ARVCF to cadherin-based cellular junctions. J Cell Sci. 2000;113:4121–35.PubMed Kaufmann U, Zuppinger C, Waibler Z, Rudiger M, Urbich C, Martin B, et al. The armadillo repeat region targets ARVCF to cadherin-based cellular junctions. J Cell Sci. 2000;113:4121–35.PubMed
107.
Zurück zum Zitat Cho K, Vaught TG, Ji H, Gu D, Papasakelariou-Yared C, Horstmann N, et al. Xenopus Kazrin interacts with ARVCF-catenin, spectrin and p190B RhoGAP, and modulates RhoA activity and epithelial integrity. J Cell Sci. 2010;123(Pt 23):4128–44.CrossRefPubMedPubMedCentral Cho K, Vaught TG, Ji H, Gu D, Papasakelariou-Yared C, Horstmann N, et al. Xenopus Kazrin interacts with ARVCF-catenin, spectrin and p190B RhoGAP, and modulates RhoA activity and epithelial integrity. J Cell Sci. 2010;123(Pt 23):4128–44.CrossRefPubMedPubMedCentral
108.
Zurück zum Zitat Fang X, Ji H, Kim SW, Park JI, Vaught TG, Anastasiadis PZ, et al. Vertebrate development requires ARVCF and p120 catenins and their interplay with RhoA and Rac. J Cell Biol. 2004;165(1):87–98.CrossRefPubMedPubMedCentral Fang X, Ji H, Kim SW, Park JI, Vaught TG, Anastasiadis PZ, et al. Vertebrate development requires ARVCF and p120 catenins and their interplay with RhoA and Rac. J Cell Biol. 2004;165(1):87–98.CrossRefPubMedPubMedCentral
109.
Zurück zum Zitat Mariner DJ, Wang J, Reynolds AB. ARVCF localizes to the nucleus and adherens junction and is mutually exclusive with p120(ctn) in E-cadherin complexes. J Cell Sci. 2000;113(Pt8):1481–90.PubMed Mariner DJ, Wang J, Reynolds AB. ARVCF localizes to the nucleus and adherens junction and is mutually exclusive with p120(ctn) in E-cadherin complexes. J Cell Sci. 2000;113(Pt8):1481–90.PubMed
110.
Zurück zum Zitat Rappe U, Schlechter T, Aschoff M, Hotz-Wagenblatt A, Hofmann I. Nuclear ARVCF protein binds splicing factors and contributes to the regulation of alternative splicing. J Biol Chem. 2014;289(18):12421–34.CrossRefPubMedPubMedCentral Rappe U, Schlechter T, Aschoff M, Hotz-Wagenblatt A, Hofmann I. Nuclear ARVCF protein binds splicing factors and contributes to the regulation of alternative splicing. J Biol Chem. 2014;289(18):12421–34.CrossRefPubMedPubMedCentral
111.
Zurück zum Zitat Ulfig N, Chan WY. Expression of ARVCF in the human ganglionic eminence during fetal development. Dev Neurosci. 2004;26(1):38–44.CrossRefPubMed Ulfig N, Chan WY. Expression of ARVCF in the human ganglionic eminence during fetal development. Dev Neurosci. 2004;26(1):38–44.CrossRefPubMed
112.
Zurück zum Zitat Cho K, Lee M, Gu D, Munoz WA, Ji H, Kloc M, et al. Kazrin, and its binding partners ARVCF- and delta- catenin, are required for Xenopus laevis craniofacial development. Dev Dyn. 2011;240(12):2601–12.CrossRefPubMedPubMedCentral Cho K, Lee M, Gu D, Munoz WA, Ji H, Kloc M, et al. Kazrin, and its binding partners ARVCF- and delta- catenin, are required for Xenopus laevis craniofacial development. Dev Dyn. 2011;240(12):2601–12.CrossRefPubMedPubMedCentral
113.
Zurück zum Zitat Tran HT, Delvaeye M, Verschuere V, Descamps E, Crabbe E, Van Hoorebeke L, et al. ARVCF depletion cooperates with Tbx1 deficiency in the development of 22q11.2DS-like phenotypes in Xenopus. Dev Dyn. 2011;240(12):2680–7.CrossRefPubMed Tran HT, Delvaeye M, Verschuere V, Descamps E, Crabbe E, Van Hoorebeke L, et al. ARVCF depletion cooperates with Tbx1 deficiency in the development of 22q11.2DS-like phenotypes in Xenopus. Dev Dyn. 2011;240(12):2680–7.CrossRefPubMed
114.
Zurück zum Zitat Marciano DK, Brakeman PR, Lee CZ, Spivak N, Eastburn DJ, Bryant DM, et al. p120 catenin is required for normal renal tubulogenesis and glomerulogenesis. Development. 2011;138(10):2099–109.CrossRefPubMedPubMedCentral Marciano DK, Brakeman PR, Lee CZ, Spivak N, Eastburn DJ, Bryant DM, et al. p120 catenin is required for normal renal tubulogenesis and glomerulogenesis. Development. 2011;138(10):2099–109.CrossRefPubMedPubMedCentral
115.
Zurück zum Zitat Mas S, Bernardo M, Parellada E, Garcia-Rizo C, Gasso P, Alvarez S, et al. ARVCF single marker and haplotypic association with schizophrenia. Prog Neuro-Psychopharmacol Biol Psychiatry. 2009;33(6):1064–9.CrossRef Mas S, Bernardo M, Parellada E, Garcia-Rizo C, Gasso P, Alvarez S, et al. ARVCF single marker and haplotypic association with schizophrenia. Prog Neuro-Psychopharmacol Biol Psychiatry. 2009;33(6):1064–9.CrossRef
116.
Zurück zum Zitat Sanders AR, Rusu I, Duan J, Vander Molen JE, Hou C, Schwab SG, et al. Haplotypic association spanning the 22q11.21 genes COMT and ARVCF with schizophrenia. Mol Psychiatry. 2005;10(4):353–65.CrossRefPubMed Sanders AR, Rusu I, Duan J, Vander Molen JE, Hou C, Schwab SG, et al. Haplotypic association spanning the 22q11.21 genes COMT and ARVCF with schizophrenia. Mol Psychiatry. 2005;10(4):353–65.CrossRefPubMed
117.
Zurück zum Zitat Furuse M, Tsukita S. Claudins in occluding junctions of humans and flies. Trends Cell Biol. 2006;16(4):181–8.CrossRefPubMed Furuse M, Tsukita S. Claudins in occluding junctions of humans and flies. Trends Cell Biol. 2006;16(4):181–8.CrossRefPubMed
118.
Zurück zum Zitat Heiskala M, Peterson PA, Yang Y. The roles of claudin superfamily proteins in paracellular transport. Traffic. 2001;2(2):93–8.CrossRefPubMed Heiskala M, Peterson PA, Yang Y. The roles of claudin superfamily proteins in paracellular transport. Traffic. 2001;2(2):93–8.CrossRefPubMed
119.
Zurück zum Zitat Krause G, Winkler L, Mueller SL, Haseloff RF, Piontek J, Blasig IE. Structure and function of claudins. Biochim Biophys Acta. 2008;1778(3):631–45.CrossRefPubMed Krause G, Winkler L, Mueller SL, Haseloff RF, Piontek J, Blasig IE. Structure and function of claudins. Biochim Biophys Acta. 2008;1778(3):631–45.CrossRefPubMed
120.
Zurück zum Zitat Capaldo CT, Nusrat A. Cytokine regulation of tight junctions. Biochim Biophys Acta. 2009;1788(4):864–71.CrossRefPubMed Capaldo CT, Nusrat A. Cytokine regulation of tight junctions. Biochim Biophys Acta. 2009;1788(4):864–71.CrossRefPubMed
121.
Zurück zum Zitat Taddei A, Giampietro C, Conti A, Orsenigo F, Breviario F, Pirazzoli V, et al. Endothelial adherens junctions control tight junctions by VE-cadherin-mediated upregulation of claudin-5. Nat Cell Biol. 2008;10(8):923–34.CrossRefPubMed Taddei A, Giampietro C, Conti A, Orsenigo F, Breviario F, Pirazzoli V, et al. Endothelial adherens junctions control tight junctions by VE-cadherin-mediated upregulation of claudin-5. Nat Cell Biol. 2008;10(8):923–34.CrossRefPubMed
122.
Zurück zum Zitat Guillemot L, Paschoud S, Pulimeno P, Foglia A, Citi S. The cytoplasmic plaque of tight junctions: a scaffolding and signalling center. Biochim Biophys Acta. 2008;1778(3):601–13.CrossRefPubMed Guillemot L, Paschoud S, Pulimeno P, Foglia A, Citi S. The cytoplasmic plaque of tight junctions: a scaffolding and signalling center. Biochim Biophys Acta. 2008;1778(3):601–13.CrossRefPubMed
123.
Zurück zum Zitat Itoh M, Furuse M, Morita K, Kubota K, Saitou M, Tsukita S. Direct binding of three tight junction- associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins. J Cell Biol. 1999;147(6):1351–63.CrossRefPubMedPubMedCentral Itoh M, Furuse M, Morita K, Kubota K, Saitou M, Tsukita S. Direct binding of three tight junction- associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins. J Cell Biol. 1999;147(6):1351–63.CrossRefPubMedPubMedCentral
124.
Zurück zum Zitat Morita KSH, Furuse M, Tsukita S. Endothelial claudin: claudin-5/TMVCF constitutes tight junction strands in endothelial cells. J Cell Biol. 1999;147(1):185–94.CrossRefPubMedPubMedCentral Morita KSH, Furuse M, Tsukita S. Endothelial claudin: claudin-5/TMVCF constitutes tight junction strands in endothelial cells. J Cell Biol. 1999;147(1):185–94.CrossRefPubMedPubMedCentral
125.
Zurück zum Zitat Morita K, Furuse M, Fujimoto K, Tsukita S. Claudin multigene family encoding four-transmembrane domain protein components of tight junction strands. Proc Natl Acad Sci U S A. 1999;96(2):511–6.CrossRefPubMedPubMedCentral Morita K, Furuse M, Fujimoto K, Tsukita S. Claudin multigene family encoding four-transmembrane domain protein components of tight junction strands. Proc Natl Acad Sci U S A. 1999;96(2):511–6.CrossRefPubMedPubMedCentral
126.
Zurück zum Zitat Yamagishi M, Ito Y, Ariizumi T, Komazaki S, Danno H, Michiue T, et al. Claudin5 genes encoding tight junction proteins are required for Xenopus heart formation. Develop Growth Differ. 2010;52(7):665–75.CrossRef Yamagishi M, Ito Y, Ariizumi T, Komazaki S, Danno H, Michiue T, et al. Claudin5 genes encoding tight junction proteins are required for Xenopus heart formation. Develop Growth Differ. 2010;52(7):665–75.CrossRef
128.
Zurück zum Zitat Delfin DA, Xu Y, Schill KE, Mays TA, Canan BD, Zang KE, et al. Sustaining cardiac claudin-5 levels prevents functional hallmarks of cardiomyopathy in a muscular dystrophy mouse model. Mol Ther. 2012;20(7):1378–83.CrossRefPubMedPubMedCentral Delfin DA, Xu Y, Schill KE, Mays TA, Canan BD, Zang KE, et al. Sustaining cardiac claudin-5 levels prevents functional hallmarks of cardiomyopathy in a muscular dystrophy mouse model. Mol Ther. 2012;20(7):1378–83.CrossRefPubMedPubMedCentral
129.
Zurück zum Zitat Sanford JL, Edwards JD, Mays TA, Gong B, Merriam AP, Rafael-Fortney JA. Claudin-5 localizes to the lateral membranes of cardiomyocytes and is altered in utrophin/dystrophin-deficient cardiomyopathic mice. J Mol Cell Cardiol. 2005;38(2):323–32.CrossRefPubMed Sanford JL, Edwards JD, Mays TA, Gong B, Merriam AP, Rafael-Fortney JA. Claudin-5 localizes to the lateral membranes of cardiomyocytes and is altered in utrophin/dystrophin-deficient cardiomyopathic mice. J Mol Cell Cardiol. 2005;38(2):323–32.CrossRefPubMed
130.
Zurück zum Zitat Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol. 2003;161(3):653–60.CrossRefPubMedPubMedCentral Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, et al. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol. 2003;161(3):653–60.CrossRefPubMedPubMedCentral
131.
Zurück zum Zitat Mays TA, Binkley PF, Lesinski A, Doshi AA, Quaile MP, Margulies KB, et al. Claudin-5 levels are reduced in human end-stage cardiomyopathy. J Mol Cell Cardiol. 2008;45(1):81–7.CrossRefPubMed Mays TA, Binkley PF, Lesinski A, Doshi AA, Quaile MP, Margulies KB, et al. Claudin-5 levels are reduced in human end-stage cardiomyopathy. J Mol Cell Cardiol. 2008;45(1):81–7.CrossRefPubMed
132.
Zurück zum Zitat Swager SA, Delfin DA, Rastogi N, Wang H, Canan BD, Fedorov VV, et al. Claudin-5 levels are reduced from multiple cell types in human failing hearts and are associated with mislocalization of ephrin-B1. Cardiovasc Pathol. 2015;24(3):160–7.CrossRefPubMed Swager SA, Delfin DA, Rastogi N, Wang H, Canan BD, Fedorov VV, et al. Claudin-5 levels are reduced from multiple cell types in human failing hearts and are associated with mislocalization of ephrin-B1. Cardiovasc Pathol. 2015;24(3):160–7.CrossRefPubMed
133.
Zurück zum Zitat Watters AK, Rom S, Hill JD, Dematatis MK, Zhou Y, Merkel SF, et al. Identification and dynamic regulation of tight junction protein expression in human neural stem cells. Stem Cells Dev. 2015;24(12):1377–89.CrossRefPubMedPubMedCentral Watters AK, Rom S, Hill JD, Dematatis MK, Zhou Y, Merkel SF, et al. Identification and dynamic regulation of tight junction protein expression in human neural stem cells. Stem Cells Dev. 2015;24(12):1377–89.CrossRefPubMedPubMedCentral
134.
Zurück zum Zitat Kajiwara K, Nagasawa H, Shimizu-Nishikawa K, Ookura T, Kimura M, Sugaya E. Cloning of SEZ-12 encoding seizure-related and membrane-bound adhesion protein. Biochem Biophys Res Commun. 1996;222(1):144–8.CrossRefPubMed Kajiwara K, Nagasawa H, Shimizu-Nishikawa K, Ookura T, Kimura M, Sugaya E. Cloning of SEZ-12 encoding seizure-related and membrane-bound adhesion protein. Biochem Biophys Res Commun. 1996;222(1):144–8.CrossRefPubMed
135.
Zurück zum Zitat Molinard-Chenu A, Dayer A. The candidate schizophrenia risk gene DGCR2 regulates early steps of corticogenesis. Biol Psychiatry. 2018;83(8):692–706.CrossRefPubMed Molinard-Chenu A, Dayer A. The candidate schizophrenia risk gene DGCR2 regulates early steps of corticogenesis. Biol Psychiatry. 2018;83(8):692–706.CrossRefPubMed
136.
Zurück zum Zitat Mugikura SI, Katoh A, Watanabe S, Kimura M, Kajiwara K. Abnormal gait, reduced locomotor activity and impaired motor coordination in Dgcr2-deficient mice. Biochem Biophys Rep. 2016;5:120–6.PubMed Mugikura SI, Katoh A, Watanabe S, Kimura M, Kajiwara K. Abnormal gait, reduced locomotor activity and impaired motor coordination in Dgcr2-deficient mice. Biochem Biophys Rep. 2016;5:120–6.PubMed
137.
Zurück zum Zitat Xu B, Roos JL, Dexheimer P, Boone B, Plummer B, Levy S, et al. Exome sequencing supports a de novo mutational paradigm for schizophrenia. Nat Genet. 2011;43(9):864–8.CrossRefPubMedPubMedCentral Xu B, Roos JL, Dexheimer P, Boone B, Plummer B, Levy S, et al. Exome sequencing supports a de novo mutational paradigm for schizophrenia. Nat Genet. 2011;43(9):864–8.CrossRefPubMedPubMedCentral
138.
Zurück zum Zitat Hartwell LH, Mortimer R, Culotti J, Culotti M. Genetic control of the cell division cycle in yeast: V. Genetic analysis of cdc mutants. Genetics. 1973;74(2):267–86.PubMedPubMedCentral Hartwell LH, Mortimer R, Culotti J, Culotti M. Genetic control of the cell division cycle in yeast: V. Genetic analysis of cdc mutants. Genetics. 1973;74(2):267–86.PubMedPubMedCentral
139.
Zurück zum Zitat Martinez C, Ware J. Mammalian septin function in hemostasis and beyond. Exp Biol Med. 2004;229:1111–9.CrossRef Martinez C, Ware J. Mammalian septin function in hemostasis and beyond. Exp Biol Med. 2004;229:1111–9.CrossRef
140.
Zurück zum Zitat Hall PA, Jung K, Hillan KJ, Russell SE. Expression profiling the human septin gene family. J Pathol. 2005;206(3):269–78.CrossRefPubMed Hall PA, Jung K, Hillan KJ, Russell SE. Expression profiling the human septin gene family. J Pathol. 2005;206(3):269–78.CrossRefPubMed
141.
Zurück zum Zitat Beites CL, Xie H, Bowser R, Trimble WS. The septin CDCrel-1 binds syntaxin and inhibits exocytosis. Nat Neurosci. 1999;2(5):434–9.CrossRefPubMed Beites CL, Xie H, Bowser R, Trimble WS. The septin CDCrel-1 binds syntaxin and inhibits exocytosis. Nat Neurosci. 1999;2(5):434–9.CrossRefPubMed
142.
Zurück zum Zitat Kinoshita A, Noda M, Kinoshita M. Differential localization of septins in the mouse brain. J Comp Neurol. 2000;428(2):223–39.CrossRefPubMed Kinoshita A, Noda M, Kinoshita M. Differential localization of septins in the mouse brain. J Comp Neurol. 2000;428(2):223–39.CrossRefPubMed
143.
Zurück zum Zitat Amin ND, Zheng YL, Kesavapany S, Kanungo J, Guszczynski T, Sihag RK, et al. Cyclin-dependent kinase 5 phosphorylation of human septin SEPT5 (hCDCrel-1) modulates exocytosis. J Neurosci. 2008;28(14):3631–43.CrossRefPubMedPubMedCentral Amin ND, Zheng YL, Kesavapany S, Kanungo J, Guszczynski T, Sihag RK, et al. Cyclin-dependent kinase 5 phosphorylation of human septin SEPT5 (hCDCrel-1) modulates exocytosis. J Neurosci. 2008;28(14):3631–43.CrossRefPubMedPubMedCentral
144.
Zurück zum Zitat Beites CL, Campbell KA, Trimble WS. The septin Sept5/CDCrel-1 competes with α-SNAP for binding to the SNARE complex. Biochem J. 2005;385(Pt2):347–53.CrossRefPubMedPubMedCentral Beites CL, Campbell KA, Trimble WS. The septin Sept5/CDCrel-1 competes with α-SNAP for binding to the SNARE complex. Biochem J. 2005;385(Pt2):347–53.CrossRefPubMedPubMedCentral
145.
Zurück zum Zitat Dong Z, Ferger B, Paterna JC, Vogel D, Furler S, Osinde M, et al. Dopamine-dependent neurodegeneration in rats induced by viral vector-mediated overexpression of the parkin target protein, CDCrel-1. Proc Natl Acad Sci U S A. 2003;100(21):12438–43.CrossRefPubMedPubMedCentral Dong Z, Ferger B, Paterna JC, Vogel D, Furler S, Osinde M, et al. Dopamine-dependent neurodegeneration in rats induced by viral vector-mediated overexpression of the parkin target protein, CDCrel-1. Proc Natl Acad Sci U S A. 2003;100(21):12438–43.CrossRefPubMedPubMedCentral
146.
Zurück zum Zitat Yang YM, Fedchyshyn MJ, Grande G, Aitoubah J, Tsang CW, Xie H, et al. Septins regulate developmental switching from microdomain to nanodomain coupling of ca(2+) influx to neurotransmitter release at a central synapse. Neuron. 2010;67(1):100–15.CrossRefPubMedPubMedCentral Yang YM, Fedchyshyn MJ, Grande G, Aitoubah J, Tsang CW, Xie H, et al. Septins regulate developmental switching from microdomain to nanodomain coupling of ca(2+) influx to neurotransmitter release at a central synapse. Neuron. 2010;67(1):100–15.CrossRefPubMedPubMedCentral
147.
Zurück zum Zitat Peng XR, Jia Z, Zhang Y, Ware J, Trimble WS. The Septin CDCrel-1 is dispensable for normal development and neurotransmitter release. Mol Cell Biol. 2002;22(1):378–87.CrossRefPubMedPubMedCentral Peng XR, Jia Z, Zhang Y, Ware J, Trimble WS. The Septin CDCrel-1 is dispensable for normal development and neurotransmitter release. Mol Cell Biol. 2002;22(1):378–87.CrossRefPubMedPubMedCentral
148.
Zurück zum Zitat Tsang CW, Fedchyshyn M, Harrison J, Xie H, Xue J, Robinson PJ, et al. Superfluous role of mammalian septins 3 and 5 in neuronal development and synaptic transmission. Mol Cell Biol. 2008;28(23):7012–29.CrossRefPubMedPubMedCentral Tsang CW, Fedchyshyn M, Harrison J, Xie H, Xue J, Robinson PJ, et al. Superfluous role of mammalian septins 3 and 5 in neuronal development and synaptic transmission. Mol Cell Biol. 2008;28(23):7012–29.CrossRefPubMedPubMedCentral
149.
Zurück zum Zitat Barr AM, Young CE, Sawada K, Trimble WS, Phillips AG, Honer WG. Abnormalities of presynaptic protein CDCrel-1 in striatum of rats reared in social isolation: relevance to neural connectivity in schizophrenia. Eur J Neurosci. 2004;20(1):303–7.CrossRefPubMed Barr AM, Young CE, Sawada K, Trimble WS, Phillips AG, Honer WG. Abnormalities of presynaptic protein CDCrel-1 in striatum of rats reared in social isolation: relevance to neural connectivity in schizophrenia. Eur J Neurosci. 2004;20(1):303–7.CrossRefPubMed
150.
Zurück zum Zitat Harper KM, Hiramoto T, Tanigaki K, Kang G, Suzuki G, Trimble W, et al. Alterations of social interaction through genetic and environmental manipulation of the 22q11.2 gene Sept5 in the mouse brain. Hum Mol Genet. 2012;21(15):3489–99.CrossRefPubMedPubMedCentral Harper KM, Hiramoto T, Tanigaki K, Kang G, Suzuki G, Trimble W, et al. Alterations of social interaction through genetic and environmental manipulation of the 22q11.2 gene Sept5 in the mouse brain. Hum Mol Genet. 2012;21(15):3489–99.CrossRefPubMedPubMedCentral
151.
Zurück zum Zitat Son JH, Kawamata H, Yoo MS, Kim DJ, Lee YK, Kim S, et al. Neurotoxicity and behavioral deficits associated with Septin 5 accumulation in dopaminergic neurons. J Neurochem. 2005;94(4):1040–53.CrossRefPubMed Son JH, Kawamata H, Yoo MS, Kim DJ, Lee YK, Kim S, et al. Neurotoxicity and behavioral deficits associated with Septin 5 accumulation in dopaminergic neurons. J Neurochem. 2005;94(4):1040–53.CrossRefPubMed
152.
Zurück zum Zitat Suzuki G, Harper KM, Hiramoto T, Sawamura T, Lee M, Kang G, et al. Sept5 deficiency exerts pleiotropic influence on affective behaviors and cognitive functions in mice. Hum Mol Genet. 2009;18(9):1652–60.CrossRefPubMedPubMedCentral Suzuki G, Harper KM, Hiramoto T, Sawamura T, Lee M, Kang G, et al. Sept5 deficiency exerts pleiotropic influence on affective behaviors and cognitive functions in mice. Hum Mol Genet. 2009;18(9):1652–60.CrossRefPubMedPubMedCentral
153.
Zurück zum Zitat Bartsch I, Sandrock K, Lanza F, Nurden P, Hainmann I, Pavlova A, et al. Deletion of human GP1BB and SEPT5 is associated with Bernard-Soulier syndrome, platelet secretion defect, polymicrogyria, and developmental delay. Thromb Haemost. 2011;106(3):475–83.CrossRefPubMed Bartsch I, Sandrock K, Lanza F, Nurden P, Hainmann I, Pavlova A, et al. Deletion of human GP1BB and SEPT5 is associated with Bernard-Soulier syndrome, platelet secretion defect, polymicrogyria, and developmental delay. Thromb Haemost. 2011;106(3):475–83.CrossRefPubMed
154.
Zurück zum Zitat Esk C, Chen CY, Johannes L, Brodsky FM. The clathrin heavy chain isoform CHC22 functions in a novel endosomal sorting step. J Cell Biol. 2010;188(1):131–44.CrossRefPubMedPubMedCentral Esk C, Chen CY, Johannes L, Brodsky FM. The clathrin heavy chain isoform CHC22 functions in a novel endosomal sorting step. J Cell Biol. 2010;188(1):131–44.CrossRefPubMedPubMedCentral
155.
Zurück zum Zitat Hood FE, Royle SJ. Functional equivalence of the clathrin heavy chains CHC17 and CHC22 in endocytosis and mitosis. J Cell Sci. 2009;122(Pt 13):2185–90.CrossRefPubMed Hood FE, Royle SJ. Functional equivalence of the clathrin heavy chains CHC17 and CHC22 in endocytosis and mitosis. J Cell Sci. 2009;122(Pt 13):2185–90.CrossRefPubMed
156.
Zurück zum Zitat Wakeham DE, Abi-Rached L, Towler MC, Wilbur JD, Parham P, Brodsky FM. Clathrin heavy and light chain isoforms originated by independent mechanisms of gene duplication during chordate evolution. Proc Natl Acad Sci U S A. 2005;102(20):7209–14.CrossRefPubMedPubMedCentral Wakeham DE, Abi-Rached L, Towler MC, Wilbur JD, Parham P, Brodsky FM. Clathrin heavy and light chain isoforms originated by independent mechanisms of gene duplication during chordate evolution. Proc Natl Acad Sci U S A. 2005;102(20):7209–14.CrossRefPubMedPubMedCentral
157.
Zurück zum Zitat Vassilopoulos S, Esk C, Hoshino S, Funke BH, Chen CY, Plocik AM, et al. A role for the CHC22 Clathrin heavy-chain isoform in human glucose metabolism. Science. 2009;324(5931):1192–6.CrossRefPubMedPubMedCentral Vassilopoulos S, Esk C, Hoshino S, Funke BH, Chen CY, Plocik AM, et al. A role for the CHC22 Clathrin heavy-chain isoform in human glucose metabolism. Science. 2009;324(5931):1192–6.CrossRefPubMedPubMedCentral
158.
Zurück zum Zitat Kedra D, Peyrard M, Fransson I, Collins JE, Dunham I, Roe BA, et al. Characterization of a second human clathrin heavy chain polypeptide gene (CLH-22) from chromosome 22q11. Hum Mol Genet. 1996;5(5):625–31.CrossRefPubMed Kedra D, Peyrard M, Fransson I, Collins JE, Dunham I, Roe BA, et al. Characterization of a second human clathrin heavy chain polypeptide gene (CLH-22) from chromosome 22q11. Hum Mol Genet. 1996;5(5):625–31.CrossRefPubMed
159.
Zurück zum Zitat Nahorski MS, Al-Gazali L, Hertecant J, Owen DJ, Borner GH, Chen YC, et al. A novel disorder reveals clathrin heavy chain-22 is essential for human pain and touch development. Brain. 2015;138(Pt 8):2147–60.CrossRefPubMedPubMedCentral Nahorski MS, Al-Gazali L, Hertecant J, Owen DJ, Borner GH, Chen YC, et al. A novel disorder reveals clathrin heavy chain-22 is essential for human pain and touch development. Brain. 2015;138(Pt 8):2147–60.CrossRefPubMedPubMedCentral
160.
161.
Zurück zum Zitat Hoshino S, Sakamoto K, Vassilopoulos S, Camus SM, Griffin CA, Esk C, et al. The CHC22 clathrin-GLUT4 transport pathway contributes to skeletal muscle regeneration. PLoS One. 2013;8(10):e77787.CrossRefPubMedPubMedCentral Hoshino S, Sakamoto K, Vassilopoulos S, Camus SM, Griffin CA, Esk C, et al. The CHC22 clathrin-GLUT4 transport pathway contributes to skeletal muscle regeneration. PLoS One. 2013;8(10):e77787.CrossRefPubMedPubMedCentral
162.
Zurück zum Zitat Towler MC, Gleeso P, Hoshino S, Rahkila P, Manalo V, Ohkoshi N, et al. Clathrin isoform CHC22, a component of neuromuscular and myotendinous junctions, binds sorting nexin 5 and has increased expression during myogenesis and muscle regeneration. Mol Biol Cell. 2004;15(7):3181–95.CrossRefPubMedPubMedCentral Towler MC, Gleeso P, Hoshino S, Rahkila P, Manalo V, Ohkoshi N, et al. Clathrin isoform CHC22, a component of neuromuscular and myotendinous junctions, binds sorting nexin 5 and has increased expression during myogenesis and muscle regeneration. Mol Biol Cell. 2004;15(7):3181–95.CrossRefPubMedPubMedCentral
163.
Zurück zum Zitat Kehlenbach RH, Assheuer R, Kehlenbach A, Becker J, Gerace L. Stimulation of nuclear export and inhibition of nuclear import by a ran mutant deficient in binding to ran-binding protein 1. J Biol Chem. 2001;276(17):14524–31.CrossRefPubMed Kehlenbach RH, Assheuer R, Kehlenbach A, Becker J, Gerace L. Stimulation of nuclear export and inhibition of nuclear import by a ran mutant deficient in binding to ran-binding protein 1. J Biol Chem. 2001;276(17):14524–31.CrossRefPubMed
165.
166.
167.
Zurück zum Zitat Ciciarello M, Roscioli E, Di Fiore B, Di Francesco L, Sobrero F, Bernard D, et al. Nuclear reformation after mitosis requires downregulation of the Ran GTPase effector RanBP1 in mammalian cells. Chromosoma. 2010;119(6):651–68.CrossRefPubMed Ciciarello M, Roscioli E, Di Fiore B, Di Francesco L, Sobrero F, Bernard D, et al. Nuclear reformation after mitosis requires downregulation of the Ran GTPase effector RanBP1 in mammalian cells. Chromosoma. 2010;119(6):651–68.CrossRefPubMed
168.
Zurück zum Zitat Fan S, Whiteman EL, Hurd TW, McIntyre JC, Dishinger JF, Liu CJ, et al. Induction of Ran GTP drives ciliogenesis. Mol Biol Cell. 2011;22(23):4539–48.CrossRefPubMedPubMedCentral Fan S, Whiteman EL, Hurd TW, McIntyre JC, Dishinger JF, Liu CJ, et al. Induction of Ran GTP drives ciliogenesis. Mol Biol Cell. 2011;22(23):4539–48.CrossRefPubMedPubMedCentral
169.
Zurück zum Zitat Maynard TM, Haskell GT, Bhasin N, Lee JM, Gassman AA, Lieberman JA, et al. RanBP1, a velocardiofacial/DiGeorge syndrome candidate gene, is expressed at sites of mesenchymal/epithelial induction. Mech Dev. 2002;111(1–2):177–80.CrossRefPubMed Maynard TM, Haskell GT, Bhasin N, Lee JM, Gassman AA, Lieberman JA, et al. RanBP1, a velocardiofacial/DiGeorge syndrome candidate gene, is expressed at sites of mesenchymal/epithelial induction. Mech Dev. 2002;111(1–2):177–80.CrossRefPubMed
170.
Zurück zum Zitat Paronett EM, Meechan DW, Karpinski BA, LaMantia AS, Maynard TM. Ranbp1, deleted in DiGeorge/ 22q11.2 deletion syndrome, is a microcephaly gene that selectively disrupts layer 2/3 cortical projection neuron generation. Cereb Cortex. 2015;25(10):3977–93.CrossRefPubMed Paronett EM, Meechan DW, Karpinski BA, LaMantia AS, Maynard TM. Ranbp1, deleted in DiGeorge/ 22q11.2 deletion syndrome, is a microcephaly gene that selectively disrupts layer 2/3 cortical projection neuron generation. Cereb Cortex. 2015;25(10):3977–93.CrossRefPubMed
171.
Zurück zum Zitat Cheong HS, Park BL, Kim EM, Park CS, Sohn JW, Kim BJ, et al. Association of RANBP1 haplotype with smooth pursuit eye movement abnormality. Am J Med Genet B Neuropsychiatr Genet. 2011;156B(1):67–71.CrossRefPubMed Cheong HS, Park BL, Kim EM, Park CS, Sohn JW, Kim BJ, et al. Association of RANBP1 haplotype with smooth pursuit eye movement abnormality. Am J Med Genet B Neuropsychiatr Genet. 2011;156B(1):67–71.CrossRefPubMed
172.
Zurück zum Zitat Wenger TL, Kao C, McDonald-McGinn DM, Zackai EH, Bailey A, Schultz RT, et al. The role of mGluR copy number variation in genetic and environmental forms of syndromic autism spectrum disorder. Sci Rep. 2016;6:19372.CrossRefPubMedPubMedCentral Wenger TL, Kao C, McDonald-McGinn DM, Zackai EH, Bailey A, Schultz RT, et al. The role of mGluR copy number variation in genetic and environmental forms of syndromic autism spectrum disorder. Sci Rep. 2016;6:19372.CrossRefPubMedPubMedCentral
173.
Zurück zum Zitat Steegmaier M, Yang B, Yoo JS, Huang B, Shen M, Yu S, Luo Y, Scheller RH. Three novel proteins of the syntaxin/SNAP-25 family. J Biol Chem. 1998;273(51):34171–9.CrossRefPubMed Steegmaier M, Yang B, Yoo JS, Huang B, Shen M, Yu S, Luo Y, Scheller RH. Three novel proteins of the syntaxin/SNAP-25 family. J Biol Chem. 1998;273(51):34171–9.CrossRefPubMed
174.
Zurück zum Zitat Hohenstein AC, Roche PA. SNAP-29 is a promiscuous syntaxin-binding SNARE. Biochem Biophys Res Commun. 2001;285(2):167–71.CrossRefPubMed Hohenstein AC, Roche PA. SNAP-29 is a promiscuous syntaxin-binding SNARE. Biochem Biophys Res Commun. 2001;285(2):167–71.CrossRefPubMed
176.
Zurück zum Zitat Morelli E, Ginefra P, Mastrodonato V, Beznoussenko GV, Rusten TE, Bilder D, et al. Multiple functions of the SNARE protein Snap29 in autophagy, endocytic, and exocytic trafficking during epithelial formation in Drosophila. Autophagy. 2014;10(12):2251–68.CrossRefPubMed Morelli E, Ginefra P, Mastrodonato V, Beznoussenko GV, Rusten TE, Bilder D, et al. Multiple functions of the SNARE protein Snap29 in autophagy, endocytic, and exocytic trafficking during epithelial formation in Drosophila. Autophagy. 2014;10(12):2251–68.CrossRefPubMed
178.
Zurück zum Zitat Wesolowski J, Caldwell V, Paumet F. A novel function for SNAP29 (synaptosomal-associated protein of 29 kDa) in mast cell phagocytosis. PLoS One. 2012;7(11):e49886.CrossRefPubMedPubMedCentral Wesolowski J, Caldwell V, Paumet F. A novel function for SNAP29 (synaptosomal-associated protein of 29 kDa) in mast cell phagocytosis. PLoS One. 2012;7(11):e49886.CrossRefPubMedPubMedCentral
179.
Zurück zum Zitat Su Q, Mochida S, Tian JH, Mehta R, Sheng ZH. SNAP-29: a general SNARE protein that inhibits SNARE disassembly and is implicated in synaptic transmission. Proc Natl Acad Sci U S A. 2001;98(24):14038–43.CrossRefPubMedPubMedCentral Su Q, Mochida S, Tian JH, Mehta R, Sheng ZH. SNAP-29: a general SNARE protein that inhibits SNARE disassembly and is implicated in synaptic transmission. Proc Natl Acad Sci U S A. 2001;98(24):14038–43.CrossRefPubMedPubMedCentral
180.
Zurück zum Zitat Pan PY, Cai Q, Lin L, Lu PH, Duan S, Sheng ZH. SNAP-29-mediated modulation of synaptic transmission in cultured hippocampal neurons. J Biol Chem. 2005;280(27):25769–79.CrossRefPubMed Pan PY, Cai Q, Lin L, Lu PH, Duan S, Sheng ZH. SNAP-29-mediated modulation of synaptic transmission in cultured hippocampal neurons. J Biol Chem. 2005;280(27):25769–79.CrossRefPubMed
181.
Zurück zum Zitat Schardt A, Brinkmann BG, Mitkovski M, Sereda MW, Werner HB, Nave KA. The SNARE protein SNAP- 29 interacts with the GTPase Rab3A: implications for membrane trafficking in myelinating glia. J Neurosci Res. 2009;87(15):3465–79.CrossRefPubMed Schardt A, Brinkmann BG, Mitkovski M, Sereda MW, Werner HB, Nave KA. The SNARE protein SNAP- 29 interacts with the GTPase Rab3A: implications for membrane trafficking in myelinating glia. J Neurosci Res. 2009;87(15):3465–79.CrossRefPubMed
182.
Zurück zum Zitat Fuchs-Telem D, Stewart H, Rapaport D, Nousbeck J, Gat A, Gini M, et al. CEDNIK syndrome results from loss-of-function mutations in SNAP29. Br J Dermatol. 2011;164(3):610–6.PubMed Fuchs-Telem D, Stewart H, Rapaport D, Nousbeck J, Gat A, Gini M, et al. CEDNIK syndrome results from loss-of-function mutations in SNAP29. Br J Dermatol. 2011;164(3):610–6.PubMed
183.
Zurück zum Zitat Sprecher E, Ishida-Yamamoto A, Mizrahi-Koren M, Rapaport D, Goldsher D, Indelman M, et al. A mutation in SNAP29, coding for a SNARE protein involved in intracellular trafficking, causes a novel neurocutaneous syndrome characterized by cerebral dysgenesis, neuropathy, ichthyosis, and palmoplantar keratoderma. Am J Hum Genet. 2005;77(2):242–51.CrossRefPubMedPubMedCentral Sprecher E, Ishida-Yamamoto A, Mizrahi-Koren M, Rapaport D, Goldsher D, Indelman M, et al. A mutation in SNAP29, coding for a SNARE protein involved in intracellular trafficking, causes a novel neurocutaneous syndrome characterized by cerebral dysgenesis, neuropathy, ichthyosis, and palmoplantar keratoderma. Am J Hum Genet. 2005;77(2):242–51.CrossRefPubMedPubMedCentral
184.
Zurück zum Zitat Schiller SA, Seebode C, Wieser GL, Goebbels S, Mobius W, Horowitz M, et al. Establishment of two mouse models for CEDNIK syndrome reveals the pivotal role of SNAP29 in epidermal differentiation. J Invest Dermatol. 2016;136(3):672–9.CrossRefPubMed Schiller SA, Seebode C, Wieser GL, Goebbels S, Mobius W, Horowitz M, et al. Establishment of two mouse models for CEDNIK syndrome reveals the pivotal role of SNAP29 in epidermal differentiation. J Invest Dermatol. 2016;136(3):672–9.CrossRefPubMed
185.
Zurück zum Zitat Delorenzi M, Speed T. An HMM model for coiled-coil domains and a comparison with PSSM-based predictions. Bioinformatics. 2002;18(4):617–25.CrossRefPubMed Delorenzi M, Speed T. An HMM model for coiled-coil domains and a comparison with PSSM-based predictions. Bioinformatics. 2002;18(4):617–25.CrossRefPubMed
186.
Zurück zum Zitat Lindsay EA, Harvey EL, Scambler PJ, Baldini A. ES2, a gene deleted in DiGeorge syndrome, encodes a nuclear protein and is expressed during early mouse development, where it shares an expression domain with a Goosecoid-like gene. Hum Mol Genet. 1998;7(4):629–35.CrossRefPubMed Lindsay EA, Harvey EL, Scambler PJ, Baldini A. ES2, a gene deleted in DiGeorge syndrome, encodes a nuclear protein and is expressed during early mouse development, where it shares an expression domain with a Goosecoid-like gene. Hum Mol Genet. 1998;7(4):629–35.CrossRefPubMed
187.
Zurück zum Zitat Noma K, Goncharov A, Jin Y. Systematic analyses of rpm-1 suppressors reveal roles for ESS-2 in mRNA splicing in Caenorhabditis elegans. Genetics. 2014;198(3):1101–15.CrossRefPubMedPubMedCentral Noma K, Goncharov A, Jin Y. Systematic analyses of rpm-1 suppressors reveal roles for ESS-2 in mRNA splicing in Caenorhabditis elegans. Genetics. 2014;198(3):1101–15.CrossRefPubMedPubMedCentral
188.
Zurück zum Zitat Rizzu P, Lindsay EA, Taylor C, O'Donnell H, Levy A, Scambler P, et al. Cloning and comparative mapping of a gene from the commonly deleted region of DiGeorge and Velocardiofacial syndromes conserved in C. elegans. Mamm Genome. 1996;7(9):639–43.CrossRefPubMed Rizzu P, Lindsay EA, Taylor C, O'Donnell H, Levy A, Scambler P, et al. Cloning and comparative mapping of a gene from the commonly deleted region of DiGeorge and Velocardiofacial syndromes conserved in C. elegans. Mamm Genome. 1996;7(9):639–43.CrossRefPubMed
189.
Zurück zum Zitat Taricani L, Tejada ML, Young PG. The fission yeast ES2 homologue, Bis1, interacts with the Ish1 stress- responsive nuclear envelope protein. J Biol Chem. 2002;277(12):10562–72.CrossRefPubMed Taricani L, Tejada ML, Young PG. The fission yeast ES2 homologue, Bis1, interacts with the Ish1 stress- responsive nuclear envelope protein. J Biol Chem. 2002;277(12):10562–72.CrossRefPubMed
190.
Zurück zum Zitat Funato H, Sato M, Sinton CM, Gautron L, Williams SC, Skach A, et al. Loss of Goosecoid-like and DiGeorge syndrome critical region 14 in interpeduncular nucleus results in altered regulation of rapid eye movement sleep. Proc Natl Acad Sci U S A. 2010;107(42):18155–60.CrossRefPubMedPubMedCentral Funato H, Sato M, Sinton CM, Gautron L, Williams SC, Skach A, et al. Loss of Goosecoid-like and DiGeorge syndrome critical region 14 in interpeduncular nucleus results in altered regulation of rapid eye movement sleep. Proc Natl Acad Sci U S A. 2010;107(42):18155–60.CrossRefPubMedPubMedCentral
191.
Zurück zum Zitat Hegele A, Kamburov A, Grossmann A, Sourlis C, Wowro S, Weimann M, et al. Dynamic protein-protein interaction wiring of the human spliceosome. Mol Cell. 2012;45(4):567–80.CrossRefPubMed Hegele A, Kamburov A, Grossmann A, Sourlis C, Wowro S, Weimann M, et al. Dynamic protein-protein interaction wiring of the human spliceosome. Mol Cell. 2012;45(4):567–80.CrossRefPubMed
192.
Zurück zum Zitat Bessonov S, Anokhina M, Will CL, Urlaub H, Lührmann R. Isolation of an active step I spliceosome and composition of its RNP core. Nature. 2008;452(7189):846–50.CrossRefPubMed Bessonov S, Anokhina M, Will CL, Urlaub H, Lührmann R. Isolation of an active step I spliceosome and composition of its RNP core. Nature. 2008;452(7189):846–50.CrossRefPubMed
193.
Zurück zum Zitat Takada I, Tsuchiya M, Yanaka K, Hidano S, Takahashi S, Kobayashi T, et al. Ess2 bridges transcriptional regulators and spliceosomal complexes via distinct interacting domains. Biochem Biophys Res Commun. 2018;497(2):597–604.CrossRefPubMed Takada I, Tsuchiya M, Yanaka K, Hidano S, Takahashi S, Kobayashi T, et al. Ess2 bridges transcriptional regulators and spliceosomal complexes via distinct interacting domains. Biochem Biophys Res Commun. 2018;497(2):597–604.CrossRefPubMed
194.
Zurück zum Zitat Takada I. DGCR14 induces Il17a gene expression through the RORgamma/BAZ1B/RSKS2 complex. Mol Cell Biol. 2015;35(2):344–55.CrossRefPubMed Takada I. DGCR14 induces Il17a gene expression through the RORgamma/BAZ1B/RSKS2 complex. Mol Cell Biol. 2015;35(2):344–55.CrossRefPubMed
195.
Zurück zum Zitat Gong W, Emaneul B, Galili N, Kim DH, Roe B, Driscoll DA, et al. Structural and mutational analysis of a conserved gene (DGSI) from the minimal DiGeorge syndrome critical region. Hum Mol Genet. 1997;6(2):267–76.CrossRefPubMed Gong W, Emaneul B, Galili N, Kim DH, Roe B, Driscoll DA, et al. Structural and mutational analysis of a conserved gene (DGSI) from the minimal DiGeorge syndrome critical region. Hum Mol Genet. 1997;6(2):267–76.CrossRefPubMed
196.
Zurück zum Zitat Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the microprocessor complex. Nature. 2004;432(7014):231–5.CrossRefPubMed Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the microprocessor complex. Nature. 2004;432(7014):231–5.CrossRefPubMed
197.
Zurück zum Zitat Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, et al. The microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432(7014):235–40.CrossRefPubMed Gregory RI, Yan KP, Amuthan G, Chendrimada T, Doratotaj B, Cooch N, et al. The microprocessor complex mediates the genesis of microRNAs. Nature. 2004;432(7014):235–40.CrossRefPubMed
198.
Zurück zum Zitat Macias S, Cordiner RA, Gautier P, Plass M, Caceres JF. DGCR8 acts as an adaptor for the exosome complex to degrade double-stranded structured RNAs. Mol Cell. 2015;60(6):873–85.CrossRefPubMedPubMedCentral Macias S, Cordiner RA, Gautier P, Plass M, Caceres JF. DGCR8 acts as an adaptor for the exosome complex to degrade double-stranded structured RNAs. Mol Cell. 2015;60(6):873–85.CrossRefPubMedPubMedCentral
199.
Zurück zum Zitat Babiarz JE, Hsu R, Melton C, Thomas M, Ullian EM, Blelloch R. A role for noncanonical microRNAs in the mammalian brain revealed by phenotypic differences in Dgcr8 versus Dicer1 knockouts and small RNA sequencing. RNA. 2011;17(8):1489–501.CrossRefPubMedPubMedCentral Babiarz JE, Hsu R, Melton C, Thomas M, Ullian EM, Blelloch R. A role for noncanonical microRNAs in the mammalian brain revealed by phenotypic differences in Dgcr8 versus Dicer1 knockouts and small RNA sequencing. RNA. 2011;17(8):1489–501.CrossRefPubMedPubMedCentral
200.
Zurück zum Zitat Stark KL, Xu B, Bagchi A, Lai WS, Liu H, Hsu R, et al. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat Genet. 2008;40(6):751–60.CrossRefPubMed Stark KL, Xu B, Bagchi A, Lai WS, Liu H, Hsu R, et al. Altered brain microRNA biogenesis contributes to phenotypic deficits in a 22q11-deletion mouse model. Nat Genet. 2008;40(6):751–60.CrossRefPubMed
201.
202.
Zurück zum Zitat Luhur A, Chawla G, Wu YC, Li J, Sokol NS. Drosha-independent DGCR8/pasha pathway regulates neuronal morphogenesis. Proc Natl Acad Sci U S A. 2014;111(4):1421–6.CrossRefPubMedPubMedCentral Luhur A, Chawla G, Wu YC, Li J, Sokol NS. Drosha-independent DGCR8/pasha pathway regulates neuronal morphogenesis. Proc Natl Acad Sci U S A. 2014;111(4):1421–6.CrossRefPubMedPubMedCentral
203.
Zurück zum Zitat Fénelon K, Mukai J, Xu B, Hsu PK, Drew LJ, Karayiorgou M, et al. Deficiency of Dgcr8, a gene disrupted by the 22q11.2 microdeletion, results in altered short-term plasticity in the prefrontal cortex. Proc Natl Acad Sci U S A. 2011;108(11):4447–52.CrossRefPubMedPubMedCentral Fénelon K, Mukai J, Xu B, Hsu PK, Drew LJ, Karayiorgou M, et al. Deficiency of Dgcr8, a gene disrupted by the 22q11.2 microdeletion, results in altered short-term plasticity in the prefrontal cortex. Proc Natl Acad Sci U S A. 2011;108(11):4447–52.CrossRefPubMedPubMedCentral
204.
Zurück zum Zitat Hsu R, Schofield CM, Dela Cruz CG, Jones-Davis DM, Blelloch R, Ullian EM. Loss of microRNAs in pyramidal neurons leads to specific changes in inhibitory synaptic transmission in the prefrontal cortex. Mol Cell Neurosci. 2012;50(3–4):283–92.CrossRefPubMedPubMedCentral Hsu R, Schofield CM, Dela Cruz CG, Jones-Davis DM, Blelloch R, Ullian EM. Loss of microRNAs in pyramidal neurons leads to specific changes in inhibitory synaptic transmission in the prefrontal cortex. Mol Cell Neurosci. 2012;50(3–4):283–92.CrossRefPubMedPubMedCentral
206.
Zurück zum Zitat Meechan DW, Tucker ES, Maynard TM, LaMantia AS. Cxcr4 regulation of interneuron migration is disrupted in 22q11.2 deletion syndrome. Proc Natl Acad Sci U S A. 2012;109(45):18601–6.CrossRefPubMedPubMedCentral Meechan DW, Tucker ES, Maynard TM, LaMantia AS. Cxcr4 regulation of interneuron migration is disrupted in 22q11.2 deletion syndrome. Proc Natl Acad Sci U S A. 2012;109(45):18601–6.CrossRefPubMedPubMedCentral
207.
Zurück zum Zitat Toritsuka M, Kimoto S, Muraki K, Landek-Salgado MA, Yoshida A, Yamamoto N, et al. Deficits in microRNA-mediated Cxcr4/Cxcl12 signaling in neurodevelopmental deficits in a 22q11 deletion syndrome mouse model. Proc Natl Acad Sci U S A. 2013;110(43):17552–7.CrossRefPubMedPubMedCentral Toritsuka M, Kimoto S, Muraki K, Landek-Salgado MA, Yoshida A, Yamamoto N, et al. Deficits in microRNA-mediated Cxcr4/Cxcl12 signaling in neurodevelopmental deficits in a 22q11 deletion syndrome mouse model. Proc Natl Acad Sci U S A. 2013;110(43):17552–7.CrossRefPubMedPubMedCentral
208.
Zurück zum Zitat Lin HP, Oksuz I, Hurley E, Wrabetz L, Awatramani R. Microprocessor complex subunit DiGeorge syndrome critical region gene 8 (Dgcr8) is required for schwann cell myelination and myelin maintenance. J Biol Chem. 2015;290(40):24294–307.CrossRefPubMedPubMedCentral Lin HP, Oksuz I, Hurley E, Wrabetz L, Awatramani R. Microprocessor complex subunit DiGeorge syndrome critical region gene 8 (Dgcr8) is required for schwann cell myelination and myelin maintenance. J Biol Chem. 2015;290(40):24294–307.CrossRefPubMedPubMedCentral
209.
Zurück zum Zitat Chapnik E, Sasson V, Blelloch R, Hornstein E. Dgcr8 controls neural crest cells survival in cardiovascular development. Dev Biol. 2012;362(1):50–6.CrossRefPubMed Chapnik E, Sasson V, Blelloch R, Hornstein E. Dgcr8 controls neural crest cells survival in cardiovascular development. Dev Biol. 2012;362(1):50–6.CrossRefPubMed
210.
Zurück zum Zitat Rao PK, Toyama Y, Chiang HR, Gupta S, Bauer M, Medvid R, et al. Loss of cardiac microRNA-mediated regulation leads to dilated cardiomyopathy and heart failure. Circ Res. 2009;105(6):585–94.CrossRefPubMedPubMedCentral Rao PK, Toyama Y, Chiang HR, Gupta S, Bauer M, Medvid R, et al. Loss of cardiac microRNA-mediated regulation leads to dilated cardiomyopathy and heart failure. Circ Res. 2009;105(6):585–94.CrossRefPubMedPubMedCentral
211.
Zurück zum Zitat Baldini A, Fulcoli FG, Illingworth E. Tbx1: transcriptional and developmental functions. Curr Top Dev Biol. 2017;122:223–43.CrossRefPubMed Baldini A, Fulcoli FG, Illingworth E. Tbx1: transcriptional and developmental functions. Curr Top Dev Biol. 2017;122:223–43.CrossRefPubMed
212.
Zurück zum Zitat Ataliotis P, Ivins S, Mohun TJ, Scambler PJ. XTbx1 is a transcriptional activator involved in head and pharyngeal arch development in Xenopus laevis. Dev Dyn. 2005;232(4):979–91.CrossRefPubMed Ataliotis P, Ivins S, Mohun TJ, Scambler PJ. XTbx1 is a transcriptional activator involved in head and pharyngeal arch development in Xenopus laevis. Dev Dyn. 2005;232(4):979–91.CrossRefPubMed
213.
Zurück zum Zitat Garg V, Yamagishi C, Hu T, Kathiriya IS, Yamagishi H, Srivastava D. Tbx1, a DiGeorge syndrome candidate gene, is regulated by sonic hedgehog during pharyngeal arch development. Dev Biol. 2001;235(1):62–73.CrossRefPubMed Garg V, Yamagishi C, Hu T, Kathiriya IS, Yamagishi H, Srivastava D. Tbx1, a DiGeorge syndrome candidate gene, is regulated by sonic hedgehog during pharyngeal arch development. Dev Biol. 2001;235(1):62–73.CrossRefPubMed
214.
Zurück zum Zitat Piotrowski T, Ahn DG, Schilling TF, Nair S, Ruvinsky I, Geisler R, et al. The zebrafish van gogh mutation disrupts tbx1, which is involved in the DiGeorge deletion syndrome in humans. Development. 2003;130(20):5043–52.CrossRefPubMed Piotrowski T, Ahn DG, Schilling TF, Nair S, Ruvinsky I, Geisler R, et al. The zebrafish van gogh mutation disrupts tbx1, which is involved in the DiGeorge deletion syndrome in humans. Development. 2003;130(20):5043–52.CrossRefPubMed
215.
Zurück zum Zitat Smith SJ, Ataliotis P, Kotecha S, Towers N, Sparrow DB, Mohun TJ. The MLC1v gene provides a transgenic marker of myocardium formation within developing chambers of the Xenopus heart. Dev Dyn. 2005;232(4):1003–12.CrossRefPubMed Smith SJ, Ataliotis P, Kotecha S, Towers N, Sparrow DB, Mohun TJ. The MLC1v gene provides a transgenic marker of myocardium formation within developing chambers of the Xenopus heart. Dev Dyn. 2005;232(4):1003–12.CrossRefPubMed
216.
Zurück zum Zitat Merscher S, Funke B, Epstein JA, Heyer J, Puech A, Lu MM, et al. TBX1 is responsible for cardiovascular defects in velo-cardio-facial/DiGeorge syndrome. Cell. 2001;104(4):619–29.CrossRefPubMed Merscher S, Funke B, Epstein JA, Heyer J, Puech A, Lu MM, et al. TBX1 is responsible for cardiovascular defects in velo-cardio-facial/DiGeorge syndrome. Cell. 2001;104(4):619–29.CrossRefPubMed
217.
Zurück zum Zitat Lindsay EA, Vitelli F, Su H, Morishima M, Huynh T, Pramparo T, et al. Tbx1 haploinsufficieny in the DiGeorge syndrome region causes aortic arch defects in mice. Nature. 2001;410(6824):97–101.CrossRefPubMed Lindsay EA, Vitelli F, Su H, Morishima M, Huynh T, Pramparo T, et al. Tbx1 haploinsufficieny in the DiGeorge syndrome region causes aortic arch defects in mice. Nature. 2001;410(6824):97–101.CrossRefPubMed
218.
Zurück zum Zitat Jerome LA, Papaioannou VE. DiGeorge syndrome phenotype in mice mutant for the T-box gene, Tbx1. Nat Genet. 2001;27(3):286–91.CrossRefPubMed Jerome LA, Papaioannou VE. DiGeorge syndrome phenotype in mice mutant for the T-box gene, Tbx1. Nat Genet. 2001;27(3):286–91.CrossRefPubMed
219.
Zurück zum Zitat Arnold JS, Werling U, Braunstein EM, Liao J, Nowotschin S, Edelmann W, et al. Inactivation of Tbx1 in the pharyngeal endoderm results in 22q11DS malformations. Development. 2006;133(5):977–87.CrossRefPubMed Arnold JS, Werling U, Braunstein EM, Liao J, Nowotschin S, Edelmann W, et al. Inactivation of Tbx1 in the pharyngeal endoderm results in 22q11DS malformations. Development. 2006;133(5):977–87.CrossRefPubMed
220.
Zurück zum Zitat Vitelli F, Morishima M, Taddei I, Lindsay EA, Baldini A. Tbx1 mutation causes multiple cardiovascular defects and disrupts neural crest and cranial nerve migratory pathways. Hum Mol Genet. 2002;11(8):915–22.CrossRefPubMed Vitelli F, Morishima M, Taddei I, Lindsay EA, Baldini A. Tbx1 mutation causes multiple cardiovascular defects and disrupts neural crest and cranial nerve migratory pathways. Hum Mol Genet. 2002;11(8):915–22.CrossRefPubMed
221.
Zurück zum Zitat Dastjerdi A, Robson L, Walker R, Hadley J, Zhang Z, Rodriguez-Niedenführ M, et al. Tbx1 regulation of myogenic differentiation in the limb and cranial mesoderm. Dev Dyn. 2007;236(2):353–63.CrossRefPubMed Dastjerdi A, Robson L, Walker R, Hadley J, Zhang Z, Rodriguez-Niedenführ M, et al. Tbx1 regulation of myogenic differentiation in the limb and cranial mesoderm. Dev Dyn. 2007;236(2):353–63.CrossRefPubMed
222.
Zurück zum Zitat Vitelli F. TBX1 is required for inner ear morphogenesis. Human molecular genetics. 2003;12(16):2041–8.CrossRefPubMed Vitelli F. TBX1 is required for inner ear morphogenesis. Human molecular genetics. 2003;12(16):2041–8.CrossRefPubMed
223.
Zurück zum Zitat Zoupa M, Seppala M, Mitsiadis T, Cobourne MT. Tbx1 is expressed at multiple sites of epithelial- mesenchymal interaction during early development of the facial complex. Int J Dev Biol. 2006;50(5):504–10.PubMed Zoupa M, Seppala M, Mitsiadis T, Cobourne MT. Tbx1 is expressed at multiple sites of epithelial- mesenchymal interaction during early development of the facial complex. Int J Dev Biol. 2006;50(5):504–10.PubMed
224.
Zurück zum Zitat Vitelli F, Taddei I, Morishima M, Meyers EN, Lindsay EA, Baldini A. A genetic link between Tbx1 and fibroblast growth factor signaling. Development. 2002;129(19):4605–11.PubMed Vitelli F, Taddei I, Morishima M, Meyers EN, Lindsay EA, Baldini A. A genetic link between Tbx1 and fibroblast growth factor signaling. Development. 2002;129(19):4605–11.PubMed
225.
Zurück zum Zitat Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, Minoshima S, et al. Role of TBX1 in human del22q11.2 syndrome. Lancet. 2003;362(9393):1366–73.CrossRefPubMed Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, Minoshima S, et al. Role of TBX1 in human del22q11.2 syndrome. Lancet. 2003;362(9393):1366–73.CrossRefPubMed
226.
Zurück zum Zitat Funke B, Saint-Jore B, Puech A, Sirotkin H, Edelmann L, Carlson C, et al. Characterization and mutation analysis of goosecoid-like (GSCL), a homeodomain-containing gene that maps to the critical region for VCFS/DGS on 22q11. Genomics. 1997;46(3):364–72.CrossRefPubMed Funke B, Saint-Jore B, Puech A, Sirotkin H, Edelmann L, Carlson C, et al. Characterization and mutation analysis of goosecoid-like (GSCL), a homeodomain-containing gene that maps to the critical region for VCFS/DGS on 22q11. Genomics. 1997;46(3):364–72.CrossRefPubMed
227.
Zurück zum Zitat Rivera-Pérez JA, Mallo M, Gendron-Maguire M, Gridley T, Behringer RR. Goosecoid is not an essential component of the mouse gastrula organizer but is required for craniofacial and rib development. Development. 1995;121(9):3005–12.PubMed Rivera-Pérez JA, Mallo M, Gendron-Maguire M, Gridley T, Behringer RR. Goosecoid is not an essential component of the mouse gastrula organizer but is required for craniofacial and rib development. Development. 1995;121(9):3005–12.PubMed
228.
Zurück zum Zitat Gottlieb S, Hanes DS, Golden JA, Oakey RJ, Budarf ML. Goosecoid-like, a gene deleted in DiGeorge and velocardiofacial syndromes, recognizes DNA with a bicoid-like specificity and is expressed in the developing mouse brain. Hum Mol Genet. 1998;7(9):1497–505.CrossRefPubMed Gottlieb S, Hanes DS, Golden JA, Oakey RJ, Budarf ML. Goosecoid-like, a gene deleted in DiGeorge and velocardiofacial syndromes, recognizes DNA with a bicoid-like specificity and is expressed in the developing mouse brain. Hum Mol Genet. 1998;7(9):1497–505.CrossRefPubMed
229.
Zurück zum Zitat Saint-Jore B, Puech A, Heyer J, Lin Q, Raine C, Kucherlapati R, Skoultchi AI. Goosecoid-like (Gscl), a candidate gene for velocardiofacial syndrome, is not essential for normal mouse development. Hum Mol Genet. 1998;7(12):1841–9.CrossRefPubMed Saint-Jore B, Puech A, Heyer J, Lin Q, Raine C, Kucherlapati R, Skoultchi AI. Goosecoid-like (Gscl), a candidate gene for velocardiofacial syndrome, is not essential for normal mouse development. Hum Mol Genet. 1998;7(12):1841–9.CrossRefPubMed
230.
Zurück zum Zitat Malik S, Roeder RG. The metazoan mediator co-activator complex as an integrative hub for transcriptional regulation. Nat Rev Genet. 2010;11(11):761–72.CrossRefPubMedPubMedCentral Malik S, Roeder RG. The metazoan mediator co-activator complex as an integrative hub for transcriptional regulation. Nat Rev Genet. 2010;11(11):761–72.CrossRefPubMedPubMedCentral
231.
232.
Zurück zum Zitat Suzuki Y, Nogi Y, Abe A, Fukasawa T. GAL11 protein, an auxiliary transcription activator for genes encoding galactose-metabolizing enzymes in Saccharomyces cerevisiae. Mol Cell Biol. 1992;12(10):4806.CrossRefPubMedPubMedCentral Suzuki Y, Nogi Y, Abe A, Fukasawa T. GAL11 protein, an auxiliary transcription activator for genes encoding galactose-metabolizing enzymes in Saccharomyces cerevisiae. Mol Cell Biol. 1992;12(10):4806.CrossRefPubMedPubMedCentral
233.
Zurück zum Zitat Thakur JK, Arthanari H, Yang F, Chau KH, Wagner G, Naar AM. Mediator subunit Gal11p/MED15 is required for fatty acid-dependent gene activation by yeast transcription factor Oaf1p. J Biol Chem. 2009;284(7):4422–8.CrossRefPubMed Thakur JK, Arthanari H, Yang F, Chau KH, Wagner G, Naar AM. Mediator subunit Gal11p/MED15 is required for fatty acid-dependent gene activation by yeast transcription factor Oaf1p. J Biol Chem. 2009;284(7):4422–8.CrossRefPubMed
234.
Zurück zum Zitat Yang F, Vought BW, Satterlee JS, Walker AK, Jim Sun ZY, Watts JL, et al. An ARC/mediator subunit required for SREBP control of cholesterol and lipid homeostasis. Nature. 2006;442(7103):700–4.CrossRefPubMed Yang F, Vought BW, Satterlee JS, Walker AK, Jim Sun ZY, Watts JL, et al. An ARC/mediator subunit required for SREBP control of cholesterol and lipid homeostasis. Nature. 2006;442(7103):700–4.CrossRefPubMed
235.
Zurück zum Zitat Hou NS, Gutschmidt A, Choi DY, Pather K, Shi X, Watts JL, et al. Activation of the endoplasmic reticulum unfolded protein response by lipid disequilibrium without disturbed proteostasis in vivo. Proc Natl Acad Sci U S A. 2014;111(22):E2271–80.CrossRefPubMedPubMedCentral Hou NS, Gutschmidt A, Choi DY, Pather K, Shi X, Watts JL, et al. Activation of the endoplasmic reticulum unfolded protein response by lipid disequilibrium without disturbed proteostasis in vivo. Proc Natl Acad Sci U S A. 2014;111(22):E2271–80.CrossRefPubMedPubMedCentral
236.
Zurück zum Zitat Taubert S, Hansen M, Van Gilst MR, Cooper SB, Yamamoto KR. The mediator subunit MDT-15 confers metabolic adaptation to ingested material. PLoS Genet. 2008;4(2):e1000021.CrossRefPubMedPubMedCentral Taubert S, Hansen M, Van Gilst MR, Cooper SB, Yamamoto KR. The mediator subunit MDT-15 confers metabolic adaptation to ingested material. PLoS Genet. 2008;4(2):e1000021.CrossRefPubMedPubMedCentral
237.
Zurück zum Zitat Terriente-Felix A, Lopez-Varea A, de Celis JF. Identification of genes affecting wing patterning through a loss-of-function mutagenesis screen and characterization of med15 function during wing development. Genetics. 2010;185(2):671–84.CrossRefPubMedPubMedCentral Terriente-Felix A, Lopez-Varea A, de Celis JF. Identification of genes affecting wing patterning through a loss-of-function mutagenesis screen and characterization of med15 function during wing development. Genetics. 2010;185(2):671–84.CrossRefPubMedPubMedCentral
238.
Zurück zum Zitat Kato Y, Habas R, Katsuyama Y, Näär AM, He X. A component of the ARC/mediator complex required for TGF beta/nodal signalling. Nature. 2002;418(6898):641–6.CrossRefPubMed Kato Y, Habas R, Katsuyama Y, Näär AM, He X. A component of the ARC/mediator complex required for TGF beta/nodal signalling. Nature. 2002;418(6898):641–6.CrossRefPubMed
240.
Zurück zum Zitat Albagli O, Dhordain P, Deweindt C, Lecocq G, Leprince D. The BTB/POZ domain: a new protein-protein interaction motif common to DNA- and actin-binding proteins. Cell Growth Differ. 1995;6(9):1193–8.PubMed Albagli O, Dhordain P, Deweindt C, Lecocq G, Leprince D. The BTB/POZ domain: a new protein-protein interaction motif common to DNA- and actin-binding proteins. Cell Growth Differ. 1995;6(9):1193–8.PubMed
241.
Zurück zum Zitat Beck J, Maerki S, Posch M, Metzger T, Persaud A, Scheel H, et al. Ubiquitylation-dependent localization of PLK1 in mitosis. Nat Cell Biol. 2013;15(4):430–9.CrossRefPubMedPubMedCentral Beck J, Maerki S, Posch M, Metzger T, Persaud A, Scheel H, et al. Ubiquitylation-dependent localization of PLK1 in mitosis. Nat Cell Biol. 2013;15(4):430–9.CrossRefPubMedPubMedCentral
243.
Zurück zum Zitat Furukawa M, He YJ, Borchers C, Xiong Y. Targeting of protein ubiquitination by BTB-Cullin 3-Roc1 ubiquitin ligases. Nat Cell Biol. 2003;5(11):1001–7.CrossRefPubMed Furukawa M, He YJ, Borchers C, Xiong Y. Targeting of protein ubiquitination by BTB-Cullin 3-Roc1 ubiquitin ligases. Nat Cell Biol. 2003;5(11):1001–7.CrossRefPubMed
244.
Zurück zum Zitat Adams J, Kelso R, Cooley L. The kelch repeat superfamily of proteins: propellers of cell function. Trends Cell Biol. 2000;10(1):17–24.CrossRefPubMed Adams J, Kelso R, Cooley L. The kelch repeat superfamily of proteins: propellers of cell function. Trends Cell Biol. 2000;10(1):17–24.CrossRefPubMed
245.
Zurück zum Zitat Yang Y, Allen E, Ding J, Wang W. Giant axonal neuropathy. Cell Mol Life Sci. 2007;64(5):601–9.CrossRefPubMed Yang Y, Allen E, Ding J, Wang W. Giant axonal neuropathy. Cell Mol Life Sci. 2007;64(5):601–9.CrossRefPubMed
246.
Zurück zum Zitat Nacak TG, Leptien K, Fellner D, Augustin HG, Kroll J. The BTB-kelch protein LZTR-1 is a novel Golgi protein that is degraded upon induction of apoptosis. J Biol Chem. 2006;281(8):5065–71.CrossRefPubMed Nacak TG, Leptien K, Fellner D, Augustin HG, Kroll J. The BTB-kelch protein LZTR-1 is a novel Golgi protein that is degraded upon induction of apoptosis. J Biol Chem. 2006;281(8):5065–71.CrossRefPubMed
247.
Zurück zum Zitat Paganini I, Chang VY, Capone GL, Vitte J, Benelli M, Barbetti L, et al. Expanding the mutational spectrum of LZTR1 in schwannomatosis. Eur J Hum Genet. 2015;23(7):963–8.CrossRefPubMed Paganini I, Chang VY, Capone GL, Vitte J, Benelli M, Barbetti L, et al. Expanding the mutational spectrum of LZTR1 in schwannomatosis. Eur J Hum Genet. 2015;23(7):963–8.CrossRefPubMed
248.
Zurück zum Zitat Piotrowski A, Xie J, Liu YF, Poplawski AB, Gomes AR, Madanecki P, et al. Germline loss-of-function mutations in LZTR1 predispose to an inherited disorder of multiple schwannomas. Nat Genet. 2014;46(2):182–7.CrossRefPubMed Piotrowski A, Xie J, Liu YF, Poplawski AB, Gomes AR, Madanecki P, et al. Germline loss-of-function mutations in LZTR1 predispose to an inherited disorder of multiple schwannomas. Nat Genet. 2014;46(2):182–7.CrossRefPubMed
249.
Zurück zum Zitat Johnston JJ, van der Smagt JJ, Rosenfeld JA, Pagnamenta AT, Alswaid A, Baker EH, et al. Autosomal recessive Noonan syndrome associated with biallelic LZTR1 variants. Genet Med. 2018; 20(10):1175–85.CrossRefPubMedPubMedCentral Johnston JJ, van der Smagt JJ, Rosenfeld JA, Pagnamenta AT, Alswaid A, Baker EH, et al. Autosomal recessive Noonan syndrome associated with biallelic LZTR1 variants. Genet Med. 2018; 20(10):1175–85.CrossRefPubMedPubMedCentral
250.
Zurück zum Zitat Matthews JM, Sunde M. Zinc fingers-folds for many occasions. IUBMB Life. 2002;54(6):35.CrossRef Matthews JM, Sunde M. Zinc fingers-folds for many occasions. IUBMB Life. 2002;54(6):35.CrossRef
251.
Zurück zum Zitat Brayer KJ, Segal DJ. Keep your fingers off my DNA: protein-protein interactions mediated by C2H2 zinc finger domains. Cell Biochem Biophys. 2008;50(3):111–31.CrossRefPubMed Brayer KJ, Segal DJ. Keep your fingers off my DNA: protein-protein interactions mediated by C2H2 zinc finger domains. Cell Biochem Biophys. 2008;50(3):111–31.CrossRefPubMed
252.
Zurück zum Zitat Hall TM. Multiple modes of RNA recognition by zinc finger proteins. Curr Opin Struct Biol. 2005;15(3):367–73.CrossRefPubMed Hall TM. Multiple modes of RNA recognition by zinc finger proteins. Curr Opin Struct Biol. 2005;15(3):367–73.CrossRefPubMed
253.
Zurück zum Zitat Guddat U, Bakken AH, Pieler T. Protein-mediated nuclear export of RNA: 5S rRNA containing small RNPs in xenopus oocytes. Cell. 1990;60(4):619–28.CrossRefPubMed Guddat U, Bakken AH, Pieler T. Protein-mediated nuclear export of RNA: 5S rRNA containing small RNPs in xenopus oocytes. Cell. 1990;60(4):619–28.CrossRefPubMed
254.
Zurück zum Zitat Theunissen O, Rudt F, Guddat U, Mentzel H, Pieler T. RNA and DNA binding zinc fingers in Xenopus TFIIIA. Cell. 1992;71(4):679–90.CrossRefPubMed Theunissen O, Rudt F, Guddat U, Mentzel H, Pieler T. RNA and DNA binding zinc fingers in Xenopus TFIIIA. Cell. 1992;71(4):679–90.CrossRefPubMed
255.
Zurück zum Zitat Grondin BBM, Aubry M. The KRAB zinc finger gene ZNF74 encodes an RNA-binding protein tightly associated with the nuclear matrix. J Biol Chem. 1996;271(26):15458–67.CrossRefPubMed Grondin BBM, Aubry M. The KRAB zinc finger gene ZNF74 encodes an RNA-binding protein tightly associated with the nuclear matrix. J Biol Chem. 1996;271(26):15458–67.CrossRefPubMed
256.
Zurück zum Zitat Grondin BCF, Bazinet M, Vincent M, Aubry M. Direct interaction of the KRAB/Cys2-His2 zinc finger protein ZNF74 with a hyperphosphorylated form of the RNA polymerase II largest subunit. J Biol Chem. 1997;272(44):27877–85.CrossRefPubMed Grondin BCF, Bazinet M, Vincent M, Aubry M. Direct interaction of the KRAB/Cys2-His2 zinc finger protein ZNF74 with a hyperphosphorylated form of the RNA polymerase II largest subunit. J Biol Chem. 1997;272(44):27877–85.CrossRefPubMed
257.
Zurück zum Zitat Dykes IM, van Bueren KL, Scambler PJ. HIC2 regulates isoform switching during maturation of the cardiovascular system. J Mol Cell Cardiol. 2018;114:29–37.CrossRefPubMedPubMedCentral Dykes IM, van Bueren KL, Scambler PJ. HIC2 regulates isoform switching during maturation of the cardiovascular system. J Mol Cell Cardiol. 2018;114:29–37.CrossRefPubMedPubMedCentral
258.
Zurück zum Zitat Dykes IM, van Bueren KL, Ashmore RJ, Floss T, Wurst W, Szumska D, et al. HIC2 is a novel dosage- dependent regulator of cardiac development located within the distal 22q11 deletion syndrome region. Circ Res. 2014;115(1):23–31.CrossRefPubMed Dykes IM, van Bueren KL, Ashmore RJ, Floss T, Wurst W, Szumska D, et al. HIC2 is a novel dosage- dependent regulator of cardiac development located within the distal 22q11 deletion syndrome region. Circ Res. 2014;115(1):23–31.CrossRefPubMed
259.
Zurück zum Zitat Roussigne M, Kossida S, Lavigne AC, Clouaire T, Ecochard V, Glories A, et al. The THAP domain: a novel protein motif with similarity to the DNA-binding domain of P element transposase. Trends Biochem Sci. 2003;28(2):66–9.CrossRefPubMed Roussigne M, Kossida S, Lavigne AC, Clouaire T, Ecochard V, Glories A, et al. The THAP domain: a novel protein motif with similarity to the DNA-binding domain of P element transposase. Trends Biochem Sci. 2003;28(2):66–9.CrossRefPubMed
260.
Zurück zum Zitat Reddy KC, Villeneuve AM. C. elegans HIM-17 links chromatin modification and competence for initiation of meiotic recombination. Cell. 2004;118(4):439–52.CrossRefPubMed Reddy KC, Villeneuve AM. C. elegans HIM-17 links chromatin modification and competence for initiation of meiotic recombination. Cell. 2004;118(4):439–52.CrossRefPubMed
261.
Zurück zum Zitat Macfarlan T, Kutney S, Altman B, Montross R, Yu J, Chakravarti D. Human THAP7 is a chromatin- associated, histone tail-binding protein that represses transcription via recruitment of HDAC3 and nuclear hormone receptor corepressor. J Biol Chem. 2005;280(8):7346–58.CrossRefPubMed Macfarlan T, Kutney S, Altman B, Montross R, Yu J, Chakravarti D. Human THAP7 is a chromatin- associated, histone tail-binding protein that represses transcription via recruitment of HDAC3 and nuclear hormone receptor corepressor. J Biol Chem. 2005;280(8):7346–58.CrossRefPubMed
262.
Zurück zum Zitat Macfarlan T, Parker JB, Nagata K, Chakravarti D. Thanatos-associated protein 7 associates with template activating factor-Ibeta and inhibits histone acetylation to repress transcription. Mol Endocrinol. 2006;20(2):335–47.CrossRefPubMed Macfarlan T, Parker JB, Nagata K, Chakravarti D. Thanatos-associated protein 7 associates with template activating factor-Ibeta and inhibits histone acetylation to repress transcription. Mol Endocrinol. 2006;20(2):335–47.CrossRefPubMed
263.
Zurück zum Zitat Hiramoto T, Kang G, Suzuki G, Satoh Y, Kucherlapati R, Watanabe Y, et al. Tbx1: identification of a 22q11.2 gene as a risk factor for autism spectrum disorder in a mouse model. Hum Mol Genet. 2011;20(24):4775–85.CrossRefPubMedPubMedCentral Hiramoto T, Kang G, Suzuki G, Satoh Y, Kucherlapati R, Watanabe Y, et al. Tbx1: identification of a 22q11.2 gene as a risk factor for autism spectrum disorder in a mouse model. Hum Mol Genet. 2011;20(24):4775–85.CrossRefPubMedPubMedCentral
264.
Zurück zum Zitat LaMantia AS, Moody SA, Maynard TM, Karpinski BA, Zohn IE, Mendelowitz D, et al. Hard to swallow: developmental biological insights into pediatric dysphagia. Dev Biol. 2016;409(2):329–42.CrossRefPubMed LaMantia AS, Moody SA, Maynard TM, Karpinski BA, Zohn IE, Mendelowitz D, et al. Hard to swallow: developmental biological insights into pediatric dysphagia. Dev Biol. 2016;409(2):329–42.CrossRefPubMed
265.
Zurück zum Zitat Schwab ME. Functions of Nogo proteins and their receptors in the nervous system. Nat Rev Neurosci. 2010;11(12):799–811.CrossRefPubMed Schwab ME. Functions of Nogo proteins and their receptors in the nervous system. Nat Rev Neurosci. 2010;11(12):799–811.CrossRefPubMed
266.
Zurück zum Zitat Fournier AEGT, Strittmatter SM. Identification of a receptor mediating Nogo-66 inhibition of axonal regeneration. Nature. 2001;409(6818):341–6.CrossRefPubMed Fournier AEGT, Strittmatter SM. Identification of a receptor mediating Nogo-66 inhibition of axonal regeneration. Nature. 2001;409(6818):341–6.CrossRefPubMed
267.
Zurück zum Zitat Huebner EA, Kim BG, Duffy PJ, Brown RH, Strittmatter SM. A multi-domain fragment of Nogo-A protein is a potent inhibitor of cortical axon regeneration via Nogo receptor 1. J Biol Chem. 2011;286(20):18026–36.CrossRefPubMedPubMedCentral Huebner EA, Kim BG, Duffy PJ, Brown RH, Strittmatter SM. A multi-domain fragment of Nogo-A protein is a potent inhibitor of cortical axon regeneration via Nogo receptor 1. J Biol Chem. 2011;286(20):18026–36.CrossRefPubMedPubMedCentral
268.
Zurück zum Zitat Kim JE, Liu BP, Park JH, Strittmatter SM. Nogo-66 receptor prevents raphespinal and rubrospinal axon regeneration and limits functional recovery from spinal cord injury. Neuron. 2004;44(3):439–51.CrossRefPubMed Kim JE, Liu BP, Park JH, Strittmatter SM. Nogo-66 receptor prevents raphespinal and rubrospinal axon regeneration and limits functional recovery from spinal cord injury. Neuron. 2004;44(3):439–51.CrossRefPubMed
269.
Zurück zum Zitat Wang KC, Koprivica V, Kim JA, Sivasankaran R, Guo Y, Neve RL, et al. Oligodendrocyte-myelin glycoprotein is a Nogo receptor ligand that inhibits neurite outgrowth. Nature. 2002;417(6892):941–4.CrossRefPubMed Wang KC, Koprivica V, Kim JA, Sivasankaran R, Guo Y, Neve RL, et al. Oligodendrocyte-myelin glycoprotein is a Nogo receptor ligand that inhibits neurite outgrowth. Nature. 2002;417(6892):941–4.CrossRefPubMed
270.
Zurück zum Zitat Merkler D, Metz G, Raineteau O, Dietz V, Schwab ME, Fouad K. Locomotor recovery in spinal cord-injured rats treated with an antibody neutralizing the myelin-associated neurite growth inhibitor Nogo-A. J Neurosci. 2001;21(10):3665–73.CrossRefPubMedPubMedCentral Merkler D, Metz G, Raineteau O, Dietz V, Schwab ME, Fouad K. Locomotor recovery in spinal cord-injured rats treated with an antibody neutralizing the myelin-associated neurite growth inhibitor Nogo-A. J Neurosci. 2001;21(10):3665–73.CrossRefPubMedPubMedCentral
271.
Zurück zum Zitat Emerick AJ, Neafsey E, Schwab ME, Kartje GL. Functional reorganization of the motor cortex in adult rats after cortical lesion and treatment with monoclonal antibody IN-1. J Neurosci. 2003;23(12):4826–30.CrossRefPubMedPubMedCentral Emerick AJ, Neafsey E, Schwab ME, Kartje GL. Functional reorganization of the motor cortex in adult rats after cortical lesion and treatment with monoclonal antibody IN-1. J Neurosci. 2003;23(12):4826–30.CrossRefPubMedPubMedCentral
272.
Zurück zum Zitat Lee H, Raiker SJ, Venkatesh K, Geary R, Robak LA, Zhang Y, et al. Synaptic function for the Nogo-66 receptor NgR1: regulation of dendritic spine morphology and activity-dependent synaptic strength. J Neurosci. 2008;28(11):2753–65.CrossRefPubMedPubMedCentral Lee H, Raiker SJ, Venkatesh K, Geary R, Robak LA, Zhang Y, et al. Synaptic function for the Nogo-66 receptor NgR1: regulation of dendritic spine morphology and activity-dependent synaptic strength. J Neurosci. 2008;28(11):2753–65.CrossRefPubMedPubMedCentral
273.
Zurück zum Zitat Peng X, Kim J, Zhou Z, Fink DJ, Mata M. Neuronal Nogo-A regulates glutamate receptor subunit expression in hippocampal neurons. J Neurochem. 2011;119(6):1183–93.CrossRefPubMedPubMedCentral Peng X, Kim J, Zhou Z, Fink DJ, Mata M. Neuronal Nogo-A regulates glutamate receptor subunit expression in hippocampal neurons. J Neurochem. 2011;119(6):1183–93.CrossRefPubMedPubMedCentral
274.
Zurück zum Zitat Wills ZP, Mandel-Brehm C, Mardinly AR, McCord AE, Giger RJ, Greenberg ME. The Nogo receptor family restricts synapse number in the developing hippocampus. Neuron. 2012;73(3):466–81.CrossRefPubMedPubMedCentral Wills ZP, Mandel-Brehm C, Mardinly AR, McCord AE, Giger RJ, Greenberg ME. The Nogo receptor family restricts synapse number in the developing hippocampus. Neuron. 2012;73(3):466–81.CrossRefPubMedPubMedCentral
276.
Zurück zum Zitat Karlén A, Karlsson T, Mattsson A, Lundströmer K, Codeluppi S, Pham TM. Nogo receptor 1 regulates formation of lasting memories. Proc Natl Acad Sci U S A. 2009;106(48):20476–81.CrossRefPubMedPubMedCentral Karlén A, Karlsson T, Mattsson A, Lundströmer K, Codeluppi S, Pham TM. Nogo receptor 1 regulates formation of lasting memories. Proc Natl Acad Sci U S A. 2009;106(48):20476–81.CrossRefPubMedPubMedCentral
277.
Zurück zum Zitat McGee AW, Yang Y, Fischer QS, Daw NW, Strittmatter SM. Experience-driven plasticity of visual cortex limited by myelin and Nogo receptor. Science. 2005;309(5744):2222–6.CrossRefPubMedPubMedCentral McGee AW, Yang Y, Fischer QS, Daw NW, Strittmatter SM. Experience-driven plasticity of visual cortex limited by myelin and Nogo receptor. Science. 2005;309(5744):2222–6.CrossRefPubMedPubMedCentral
278.
Zurück zum Zitat Stephany CE, Chan LL, Parivash SN, Dorton HM, Piechowicz M, Qiu S, et al. Plasticity of binocularity and visual acuity are differentially limited by nogo receptor. J Neurosci. 2014;34(35):11631–40.CrossRefPubMedPubMedCentral Stephany CE, Chan LL, Parivash SN, Dorton HM, Piechowicz M, Qiu S, et al. Plasticity of binocularity and visual acuity are differentially limited by nogo receptor. J Neurosci. 2014;34(35):11631–40.CrossRefPubMedPubMedCentral
279.
Zurück zum Zitat Lee JK, Chan AF, Luu SM, Zhu Y, Ho C, Tessier-Lavigne M, et al. Reassessment of corticospinal tract regeneration in Nogo-deficient mice. J Neurosci. 2009;29(27):8649–54.CrossRefPubMedPubMedCentral Lee JK, Chan AF, Luu SM, Zhu Y, Ho C, Tessier-Lavigne M, et al. Reassessment of corticospinal tract regeneration in Nogo-deficient mice. J Neurosci. 2009;29(27):8649–54.CrossRefPubMedPubMedCentral
280.
Zurück zum Zitat Zheng B, Atwal J, Ho C, Case L, He XL, Garcia KC, et al. Genetic deletion of the Nogo receptor does not reduce neurite inhibition in vitro or promote corticospinal tract regeneration in vivo. Proc Natl Acad Sci U S A. 2005;102(4):1205–10.CrossRefPubMedPubMedCentral Zheng B, Atwal J, Ho C, Case L, He XL, Garcia KC, et al. Genetic deletion of the Nogo receptor does not reduce neurite inhibition in vitro or promote corticospinal tract regeneration in vivo. Proc Natl Acad Sci U S A. 2005;102(4):1205–10.CrossRefPubMedPubMedCentral
281.
Zurück zum Zitat Zheng B, Ho C, Li S, Keirstead H, Steward O, Tessier-Lavigne M. Lack of enhanced spinal regeneration in Nogo-deficient mice. Neuron. 2003;38:213–24.CrossRefPubMed Zheng B, Ho C, Li S, Keirstead H, Steward O, Tessier-Lavigne M. Lack of enhanced spinal regeneration in Nogo-deficient mice. Neuron. 2003;38:213–24.CrossRefPubMed
282.
Zurück zum Zitat Mathis C, Schroter A, Thallmair M, Schwab ME. Nogo-a regulates neural precursor migration in the embryonic mouse cortex. Cereb Cortex. 2010;20(10):2380–90.CrossRefPubMedPubMedCentral Mathis C, Schroter A, Thallmair M, Schwab ME. Nogo-a regulates neural precursor migration in the embryonic mouse cortex. Cereb Cortex. 2010;20(10):2380–90.CrossRefPubMedPubMedCentral
283.
Zurück zum Zitat Ramasamy S, Yu F, Hong Yu Y, Srivats H, Dawe GS, Ahmed S. NogoR1 and PirB signaling stimulates neural stem cell survival and proliferation. Stem Cells. 2014;32(6):1636–48.CrossRefPubMed Ramasamy S, Yu F, Hong Yu Y, Srivats H, Dawe GS, Ahmed S. NogoR1 and PirB signaling stimulates neural stem cell survival and proliferation. Stem Cells. 2014;32(6):1636–48.CrossRefPubMed
284.
Zurück zum Zitat Wang B, Xiao Z, Chen B, Han J, Gao Y, Zhang J, et al. Nogo-66 promotes the differentiation of neural progenitors into astroglial lineage cells through mTOR-STAT3 pathway. PLoS One. 2008;3(3):e1856.CrossRefPubMedPubMedCentral Wang B, Xiao Z, Chen B, Han J, Gao Y, Zhang J, et al. Nogo-66 promotes the differentiation of neural progenitors into astroglial lineage cells through mTOR-STAT3 pathway. PLoS One. 2008;3(3):e1856.CrossRefPubMedPubMedCentral
285.
Zurück zum Zitat Gao Y, Wang B, Xiao Z, Chen B, Han J, Wang X, et al. Nogo-66 regulates nanog expression through stat3 pathway in murine embryonic stem cells. Stem Cells Dev. 2010;19(1):53–60.CrossRefPubMed Gao Y, Wang B, Xiao Z, Chen B, Han J, Wang X, et al. Nogo-66 regulates nanog expression through stat3 pathway in murine embryonic stem cells. Stem Cells Dev. 2010;19(1):53–60.CrossRefPubMed
286.
Zurück zum Zitat Rolando C, Parolisi R, Boda E, Schwab ME, Rossi F, Buffo A. Distinct roles of Nogo-a and Nogo receptor 1 in the homeostatic regulation of adult neural stem cell function and neuroblast migration. J Neurosci. 2012;32(49):17788–99.CrossRefPubMedPubMedCentral Rolando C, Parolisi R, Boda E, Schwab ME, Rossi F, Buffo A. Distinct roles of Nogo-a and Nogo receptor 1 in the homeostatic regulation of adult neural stem cell function and neuroblast migration. J Neurosci. 2012;32(49):17788–99.CrossRefPubMedPubMedCentral
287.
Zurück zum Zitat Perlstein MD, Chohan MR, Coman IL, Antshel KM, Fremont WP, Gnirke MH, et al. White matter abnormalities in 22q11.2 deletion syndrome: preliminary associations with the Nogo-66 receptor gene and symptoms of psychosis. Schizophr Res. 2014;152(1):117–23.CrossRefPubMed Perlstein MD, Chohan MR, Coman IL, Antshel KM, Fremont WP, Gnirke MH, et al. White matter abnormalities in 22q11.2 deletion syndrome: preliminary associations with the Nogo-66 receptor gene and symptoms of psychosis. Schizophr Res. 2014;152(1):117–23.CrossRefPubMed
288.
Zurück zum Zitat Thompson CA, Karelis J, Middleton FA, Gentile K, Coman IL, Radoeva PD, et al. Associations between neurodevelopmental genes, neuroanatomy, and ultra high risk symptoms of psychosis in 22q11.2 deletion syndrome. Am J Med Genet B Neuropsychiatr Genet. 2017;174(3):295–314.CrossRefPubMed Thompson CA, Karelis J, Middleton FA, Gentile K, Coman IL, Radoeva PD, et al. Associations between neurodevelopmental genes, neuroanatomy, and ultra high risk symptoms of psychosis in 22q11.2 deletion syndrome. Am J Med Genet B Neuropsychiatr Genet. 2017;174(3):295–314.CrossRefPubMed
289.
Zurück zum Zitat Fang Y, Yan J, Li C, Zhou X, Yao L, Pang T, et al. The Nogo/Nogo receptor (NgR) signal is involved in neuroinflammation through the regulation of microglial inflammatory activation. J Biol Chem. 2015;290(48):28901–14.CrossRefPubMedPubMedCentral Fang Y, Yan J, Li C, Zhou X, Yao L, Pang T, et al. The Nogo/Nogo receptor (NgR) signal is involved in neuroinflammation through the regulation of microglial inflammatory activation. J Biol Chem. 2015;290(48):28901–14.CrossRefPubMedPubMedCentral
290.
Zurück zum Zitat Pool M, Niino M, Rambaldi I, Robson K, Bar-Or A, Fournier AE. Myelin regulates immune cell adhesion and motility. Exp Neurol. 2009;217(2):371–7.CrossRefPubMed Pool M, Niino M, Rambaldi I, Robson K, Bar-Or A, Fournier AE. Myelin regulates immune cell adhesion and motility. Exp Neurol. 2009;217(2):371–7.CrossRefPubMed
291.
Zurück zum Zitat Yan J, Zhou X, Guo JJ, Mao L, Wang YJ, Sun J, et al. Nogo-66 inhibits adhesion and migration of microglia via GTPase rho pathway in vitro. J Neurochem. 2012;120(5):721–31.CrossRefPubMed Yan J, Zhou X, Guo JJ, Mao L, Wang YJ, Sun J, et al. Nogo-66 inhibits adhesion and migration of microglia via GTPase rho pathway in vitro. J Neurochem. 2012;120(5):721–31.CrossRefPubMed
292.
Zurück zum Zitat Evans RJ, Surprenant A. Vasoconstriction of Guinea-pig submucosal arterioles following sympathetic nerve stimulation is mediated by the release of ATP. Br J Pharmacol. 1992;106(2):242–9.CrossRefPubMedPubMedCentral Evans RJ, Surprenant A. Vasoconstriction of Guinea-pig submucosal arterioles following sympathetic nerve stimulation is mediated by the release of ATP. Br J Pharmacol. 1992;106(2):242–9.CrossRefPubMedPubMedCentral
293.
Zurück zum Zitat Nieber K, Poelchen W, Illes P. Role of ATP in fast excitatory synaptic potentials in locus coeruleus neurones of the rat. Br J Pharmacol. 1997;122(3):423–30.CrossRefPubMedPubMedCentral Nieber K, Poelchen W, Illes P. Role of ATP in fast excitatory synaptic potentials in locus coeruleus neurones of the rat. Br J Pharmacol. 1997;122(3):423–30.CrossRefPubMedPubMedCentral
294.
Zurück zum Zitat Pankratov Y, Castro E, Miras-Portugal MT, Krishtal O. A purinergic component of the excitatory post- synaptic current mediated by P2X receptors in the CA1 neurons of the rat hippocampus. Eur J Neurosci. 1998;10(12):3898–902.CrossRefPubMed Pankratov Y, Castro E, Miras-Portugal MT, Krishtal O. A purinergic component of the excitatory post- synaptic current mediated by P2X receptors in the CA1 neurons of the rat hippocampus. Eur J Neurosci. 1998;10(12):3898–902.CrossRefPubMed
295.
Zurück zum Zitat Ryten M, Hoebertz A, Burnstock G. Sequential expression of three receptor subtypes for extracellular ATP in developing rat skeletal muscle. Dev Dyn. 2001;22(3):331–41.CrossRef Ryten M, Hoebertz A, Burnstock G. Sequential expression of three receptor subtypes for extracellular ATP in developing rat skeletal muscle. Dev Dyn. 2001;22(3):331–41.CrossRef
296.
Zurück zum Zitat King BF, Towsend-Nicholson A, Wildman SS, Thomas T, Spyer KM, Burnstock G. Coexpression of rat P2X2 and P2X6 subunits in Xenopus oocytes. J Neurosci. 2000;20(13):4871–7.CrossRefPubMedPubMedCentral King BF, Towsend-Nicholson A, Wildman SS, Thomas T, Spyer KM, Burnstock G. Coexpression of rat P2X2 and P2X6 subunits in Xenopus oocytes. J Neurosci. 2000;20(13):4871–7.CrossRefPubMedPubMedCentral
297.
Zurück zum Zitat Lê KT, Babinski K, Séguéla P. Central P2X4 and P2X6 channel subunits coassemble into a novel heteromeric ATP receptor. J Neurosci. 1998;18(18):7152–9.CrossRefPubMedPubMedCentral Lê KT, Babinski K, Séguéla P. Central P2X4 and P2X6 channel subunits coassemble into a novel heteromeric ATP receptor. J Neurosci. 1998;18(18):7152–9.CrossRefPubMedPubMedCentral
298.
Zurück zum Zitat Collo G, North RA, Kawashima E, Merlo-Pich E, Neidhart S, Surprenant A, et al. Cloning of P2X, and P2X, receptors and the distribution and properties of an extended family of ATP-gated ion channels. J Neurosci. 1996;16(8):2495–507.CrossRefPubMedPubMedCentral Collo G, North RA, Kawashima E, Merlo-Pich E, Neidhart S, Surprenant A, et al. Cloning of P2X, and P2X, receptors and the distribution and properties of an extended family of ATP-gated ion channels. J Neurosci. 1996;16(8):2495–507.CrossRefPubMedPubMedCentral
299.
Zurück zum Zitat Kameda Y, Saitoh T, Nemoto N, Katoh T, Iseki S. Hes1 is required for the development of the superior cervical ganglion of sympathetic trunk and the carotid body. Dev Dyn. 2012;241(8):1289–300.CrossRefPubMed Kameda Y, Saitoh T, Nemoto N, Katoh T, Iseki S. Hes1 is required for the development of the superior cervical ganglion of sympathetic trunk and the carotid body. Dev Dyn. 2012;241(8):1289–300.CrossRefPubMed
300.
Zurück zum Zitat Cheung KK, Chan WY, Burnstock G. Expression of P2X purinoceptors during rat brain development and their inhibitory role on motor axon outgrowth in neural tube explant cultures. Neuroscience. 2005;133(4):937–45.CrossRefPubMed Cheung KK, Chan WY, Burnstock G. Expression of P2X purinoceptors during rat brain development and their inhibitory role on motor axon outgrowth in neural tube explant cultures. Neuroscience. 2005;133(4):937–45.CrossRefPubMed
301.
Zurück zum Zitat da Silva RL, Resende RR, Ulrich H. Alternative splicing of P2X6 receptors in developing mouse brain and during in vitro neuronal differentiation. Exp Physiol. 2007;92(1):139–45.PubMed da Silva RL, Resende RR, Ulrich H. Alternative splicing of P2X6 receptors in developing mouse brain and during in vitro neuronal differentiation. Exp Physiol. 2007;92(1):139–45.PubMed
302.
303.
Zurück zum Zitat Schwindt TT, Trujillo CA, Negraes PD, Lameu C, Ulrich H. Directed differentiation of neural progenitors into neurons is accompanied by altered expression of P2X purinergic receptors. J Mol Neurosci. 2011;44(3):141–6.CrossRefPubMed Schwindt TT, Trujillo CA, Negraes PD, Lameu C, Ulrich H. Directed differentiation of neural progenitors into neurons is accompanied by altered expression of P2X purinergic receptors. J Mol Neurosci. 2011;44(3):141–6.CrossRefPubMed
304.
Zurück zum Zitat Closs EI, Boissel JP, Habermeier A, Rotmann A. Structure and function of cationic amino acid transporters (CATs). J Membr Biol. 2006;213(2):67–77.CrossRefPubMed Closs EI, Boissel JP, Habermeier A, Rotmann A. Structure and function of cationic amino acid transporters (CATs). J Membr Biol. 2006;213(2):67–77.CrossRefPubMed
305.
Zurück zum Zitat Wolf SJA, Vékony N, Martiné U, Strand D, Closs EI. Expression of solute carrier 7A4 (SLC7A4) in the plasma membrane is not sufficient to mediate amino acid transport activity. Biochem J. 2002;364(Pt3):767–75.CrossRefPubMedPubMedCentral Wolf SJA, Vékony N, Martiné U, Strand D, Closs EI. Expression of solute carrier 7A4 (SLC7A4) in the plasma membrane is not sufficient to mediate amino acid transport activity. Biochem J. 2002;364(Pt3):767–75.CrossRefPubMedPubMedCentral
306.
Zurück zum Zitat Zheng B, Ho C, Li S, Keirstead H, Steward O, Tessier-Lavigne M. Lack of enhanced spinal regeneration in Nogo-deficient mice. Neuron. 2003;38(2):213–24.CrossRefPubMed Zheng B, Ho C, Li S, Keirstead H, Steward O, Tessier-Lavigne M. Lack of enhanced spinal regeneration in Nogo-deficient mice. Neuron. 2003;38(2):213–24.CrossRefPubMed
307.
308.
Zurück zum Zitat Wang SS, Brandriss MC. Proline utilization in Saccharomyces cerevisiae: sequence, regulation, and mitochondrial localization of the PUT1 gene product. Mol Cell Biol. 1987;7(12):4431–40.CrossRefPubMedPubMedCentral Wang SS, Brandriss MC. Proline utilization in Saccharomyces cerevisiae: sequence, regulation, and mitochondrial localization of the PUT1 gene product. Mol Cell Biol. 1987;7(12):4431–40.CrossRefPubMedPubMedCentral
309.
Zurück zum Zitat Takemoto Y, Semba R. Immunohistochemical evidence for the localization of neurons containing the putative transmitter L-proline in rat brain. Brain Res. 2006;1073–1074:311–5.CrossRefPubMed Takemoto Y, Semba R. Immunohistochemical evidence for the localization of neurons containing the putative transmitter L-proline in rat brain. Brain Res. 2006;1073–1074:311–5.CrossRefPubMed
310.
Zurück zum Zitat Maynard TM, Meechan DW, Dudevoir ML, Gopalakrishna D, Peters AZ, Heindel CC, et al. Mitochondrial localization and function of a subset of 22q11 deletion syndrome candidate genes. Mol Cell Neurosci. 2008;39(3):439–51.CrossRefPubMedPubMedCentral Maynard TM, Meechan DW, Dudevoir ML, Gopalakrishna D, Peters AZ, Heindel CC, et al. Mitochondrial localization and function of a subset of 22q11 deletion syndrome candidate genes. Mol Cell Neurosci. 2008;39(3):439–51.CrossRefPubMedPubMedCentral
311.
Zurück zum Zitat Meechan DW, Maynard TM, Wu Y, Gopalakrishna D, Lieberman JA, LaMantia AS. Gene dosage in the developing and adult brain in a mouse model of 22q11 deletion syndrome. Mol cell Neurosci. 2006;33(4):412-28. 312. Nadler JV, Wang A, Hakim A. toxicity of L-proline toward rat hippocampal neurons. Brain Res. 1988;456(1):168–72.CrossRef Meechan DW, Maynard TM, Wu Y, Gopalakrishna D, Lieberman JA, LaMantia AS. Gene dosage in the developing and adult brain in a mouse model of 22q11 deletion syndrome. Mol cell Neurosci. 2006;33(4):412-28. 312. Nadler JV, Wang A, Hakim A. toxicity of L-proline toward rat hippocampal neurons. Brain Res. 1988;456(1):168–72.CrossRef
312.
Zurück zum Zitat Nadler JV, Wang A, Hakim A. Toxicity of L-proline toward rat hippocampal neurons. Brain Res. 1988;456(1):168–72.CrossRefPubMed Nadler JV, Wang A, Hakim A. Toxicity of L-proline toward rat hippocampal neurons. Brain Res. 1988;456(1):168–72.CrossRefPubMed
313.
Zurück zum Zitat Nickolson VJ. “On” and “off” responses of K+-induced synaptosomal proline release: involvement of the sodium pump. J Neurochem. 1982;38(1):289–92.CrossRefPubMed Nickolson VJ. “On” and “off” responses of K+-induced synaptosomal proline release: involvement of the sodium pump. J Neurochem. 1982;38(1):289–92.CrossRefPubMed
314.
Zurück zum Zitat Ault B, Wang CM, Yawn BC. L-proline depolarizes rat spinal motoneurones by an excitatory amino acid antagonist-sensitive mechanism. Br J Pharmacol. 1987;92(2):319–26.CrossRefPubMedPubMedCentral Ault B, Wang CM, Yawn BC. L-proline depolarizes rat spinal motoneurones by an excitatory amino acid antagonist-sensitive mechanism. Br J Pharmacol. 1987;92(2):319–26.CrossRefPubMedPubMedCentral
315.
Zurück zum Zitat Cohen SM, Nadler JV. Proline-induced potentiation of glutamate transmission. Brain Res. 1997;761(2):271–82.CrossRefPubMed Cohen SM, Nadler JV. Proline-induced potentiation of glutamate transmission. Brain Res. 1997;761(2):271–82.CrossRefPubMed
316.
Zurück zum Zitat Henzi V, Reichling DB, Helm SW, MacDermott AB. L-proline activates glutamate and glycine receptors in cultured rat dorsal horn neurons. Mol Pharmacol. 1992;4(14):793–801. Henzi V, Reichling DB, Helm SW, MacDermott AB. L-proline activates glutamate and glycine receptors in cultured rat dorsal horn neurons. Mol Pharmacol. 1992;4(14):793–801.
317.
Zurück zum Zitat Martin D, Ault B, Nadler JV. NMDA receptor-mediated depolarizing action of proline on CA1 pyramidal cells. Eur J Pharmacol. 1992;219(1):59–66.CrossRefPubMed Martin D, Ault B, Nadler JV. NMDA receptor-mediated depolarizing action of proline on CA1 pyramidal cells. Eur J Pharmacol. 1992;219(1):59–66.CrossRefPubMed
318.
Zurück zum Zitat Cohen SM, Nadler JV. Proline-induced inhibition of glutamate release in hippocampal area CA1. Brain Res. 1997;769(2):333–9.CrossRefPubMed Cohen SM, Nadler JV. Proline-induced inhibition of glutamate release in hippocampal area CA1. Brain Res. 1997;769(2):333–9.CrossRefPubMed
319.
Zurück zum Zitat Bavaresco CS, Luíz Streck E, Netto CA, de Wyse ATS. Chronic hyperprolinemia provokes a memory deficit in the Morris water aaze task. Metab Brain Dis. 2005;20(1):73–80.CrossRefPubMed Bavaresco CS, Luíz Streck E, Netto CA, de Wyse ATS. Chronic hyperprolinemia provokes a memory deficit in the Morris water aaze task. Metab Brain Dis. 2005;20(1):73–80.CrossRefPubMed
320.
Zurück zum Zitat Baxter CF, Baldwin RA, Davis JL, Flood JF. High proline levels in the brains of mice as related to specific learning deficits. Pharmacol Biochem Behav. 1985;22(6):1053–9.CrossRefPubMed Baxter CF, Baldwin RA, Davis JL, Flood JF. High proline levels in the brains of mice as related to specific learning deficits. Pharmacol Biochem Behav. 1985;22(6):1053–9.CrossRefPubMed
321.
Zurück zum Zitat Davis JL, Pico RM, Flood JF. Differences in learning between hyperprolinemic mice and their congenic controls. Behav Neural Biol. 1987;48(1):128–37.CrossRefPubMed Davis JL, Pico RM, Flood JF. Differences in learning between hyperprolinemic mice and their congenic controls. Behav Neural Biol. 1987;48(1):128–37.CrossRefPubMed
322.
Zurück zum Zitat Gogos JA, Santha M, Takacs Z, Beck KD, Luine V, Lucas LR, et al. The gene encoding proline dehydro- genase modulates sensorimotor gating in mice. Nat Genet. 1999;21(4):434–9.CrossRefPubMed Gogos JA, Santha M, Takacs Z, Beck KD, Luine V, Lucas LR, et al. The gene encoding proline dehydro- genase modulates sensorimotor gating in mice. Nat Genet. 1999;21(4):434–9.CrossRefPubMed
323.
Zurück zum Zitat Bender HU, Almashanu S, Steel G, Hu CA, Lin WW, Willis A, et al. Functional consequences of PRODH missense mutations. Am J Hum Genet. 2005;76(3):409–20.CrossRefPubMedPubMedCentral Bender HU, Almashanu S, Steel G, Hu CA, Lin WW, Willis A, et al. Functional consequences of PRODH missense mutations. Am J Hum Genet. 2005;76(3):409–20.CrossRefPubMedPubMedCentral
324.
Zurück zum Zitat Afenjar A, Moutard ML, Doummar D, Guet A, Rabier D, Vermersch AI, et al. Early neurological phenotype in 4 children with biallelic PRODH mutations. Brain and Development. 2007;29(9):547–52.CrossRefPubMed Afenjar A, Moutard ML, Doummar D, Guet A, Rabier D, Vermersch AI, et al. Early neurological phenotype in 4 children with biallelic PRODH mutations. Brain and Development. 2007;29(9):547–52.CrossRefPubMed
325.
Zurück zum Zitat Jacquet H, Berthelot J, Bonnemains C, Simard G, Saugier-Veber P, Raux G, et al. The severe form of type I hyperprolinaemia results from homozygous inactivation of the PRODH gene. J Med Genet. 2003;40(1):e7.CrossRefPubMedPubMedCentral Jacquet H, Berthelot J, Bonnemains C, Simard G, Saugier-Veber P, Raux G, et al. The severe form of type I hyperprolinaemia results from homozygous inactivation of the PRODH gene. J Med Genet. 2003;40(1):e7.CrossRefPubMedPubMedCentral
326.
Zurück zum Zitat Jacquet H, Raux G, Thibaut F, Hecketsweiler B, Houy E, Demilly C, et al. PRODH mutations and hyper- prolinemia in a subset of schizophrenic patients. Hum Mol Genet. 2002;11(19):2243–9.CrossRefPubMed Jacquet H, Raux G, Thibaut F, Hecketsweiler B, Houy E, Demilly C, et al. PRODH mutations and hyper- prolinemia in a subset of schizophrenic patients. Hum Mol Genet. 2002;11(19):2243–9.CrossRefPubMed
327.
Zurück zum Zitat Johnson EC, Border R, Melroy-Greif WE, de Leeuw CA, Ehringer MA, Keller MC. No evidence that schizophrenia candidate genes are more associated with schizophrenia than noncandidate genes. Biol Psychiatry. 2017;82(10):702–8.CrossRefPubMedPubMedCentral Johnson EC, Border R, Melroy-Greif WE, de Leeuw CA, Ehringer MA, Keller MC. No evidence that schizophrenia candidate genes are more associated with schizophrenia than noncandidate genes. Biol Psychiatry. 2017;82(10):702–8.CrossRefPubMedPubMedCentral
328.
Zurück zum Zitat Jia L, Kaur J, Stuart RA. Mapping of the Saccharomyces cerevisiae Oxa1-mitochondrial ribosome interface and identification of MrpL40, a ribosomal protein in close proximity to Oxa1 and critical for oxidative phosphorylation complex assembly. Eukaryot Cell. 2009;8(11):1792–802.CrossRefPubMedPubMedCentral Jia L, Kaur J, Stuart RA. Mapping of the Saccharomyces cerevisiae Oxa1-mitochondrial ribosome interface and identification of MrpL40, a ribosomal protein in close proximity to Oxa1 and critical for oxidative phosphorylation complex assembly. Eukaryot Cell. 2009;8(11):1792–802.CrossRefPubMedPubMedCentral
329.
Zurück zum Zitat Funke BPA, Saint-Jore B, Pandita R, Skoultchi A, Morrow B. Isolation and characterization of a human gene containing a nuclear localization signal from the critical region for Velo–cardio–facial syndrome on 22q11. Genomics. 1998;53(2):146–54.CrossRefPubMed Funke BPA, Saint-Jore B, Pandita R, Skoultchi A, Morrow B. Isolation and characterization of a human gene containing a nuclear localization signal from the critical region for Velo–cardio–facial syndrome on 22q11. Genomics. 1998;53(2):146–54.CrossRefPubMed
330.
Zurück zum Zitat Allendoerfer KL, Shatz CJ. The subplate, a transient neocortical structure: its role in the development of connections between thalamus and cortex. Annu Rev Neurosci. 1994;17:185–218.CrossRefPubMed Allendoerfer KL, Shatz CJ. The subplate, a transient neocortical structure: its role in the development of connections between thalamus and cortex. Annu Rev Neurosci. 1994;17:185–218.CrossRefPubMed
331.
Zurück zum Zitat Hoerder-Suabedissen A, Molnar Z. Development, evolution and pathology of neocortical subplate neurons. Nat Rev Neurosci. 2015;16(3):133–46.CrossRefPubMed Hoerder-Suabedissen A, Molnar Z. Development, evolution and pathology of neocortical subplate neurons. Nat Rev Neurosci. 2015;16(3):133–46.CrossRefPubMed
332.
Zurück zum Zitat Devaraju P, Yu J, Eddins D, Mellado-Lagarde MM, Earls LR, Westmoreland JJ, et al. Haploinsufficiency of the 22q11.2 microdeletion gene Mrpl40 disrupts short-term synaptic plasticity and working memory through dysregulation of mitochondrial calcium. Mol Psychiatry. 2016;22(9):1313–26.CrossRefPubMedPubMedCentral Devaraju P, Yu J, Eddins D, Mellado-Lagarde MM, Earls LR, Westmoreland JJ, et al. Haploinsufficiency of the 22q11.2 microdeletion gene Mrpl40 disrupts short-term synaptic plasticity and working memory through dysregulation of mitochondrial calcium. Mol Psychiatry. 2016;22(9):1313–26.CrossRefPubMedPubMedCentral
333.
Zurück zum Zitat Bard F, Casano L, Mallabiabarrena A, Wallace E, Saito K, Kitayama H, et al. Functional genomics reveals genes involved in protein secretion and Golgi organization. Nature. 2006;439(7076):604–7.CrossRefPubMed Bard F, Casano L, Mallabiabarrena A, Wallace E, Saito K, Kitayama H, et al. Functional genomics reveals genes involved in protein secretion and Golgi organization. Nature. 2006;439(7076):604–7.CrossRefPubMed
334.
Zurück zum Zitat Kremer LS, Distelmaier F, Alhaddad B, Hempel M, Iuso A, Kupper C, et al. Bi-allelic truncating mutations in TANGO2 cause infancy-onset recurrent metabolic crises with encephalocardiomyopathy. Am J Hum Genet. 2016;98(2):358–62.CrossRefPubMedPubMedCentral Kremer LS, Distelmaier F, Alhaddad B, Hempel M, Iuso A, Kupper C, et al. Bi-allelic truncating mutations in TANGO2 cause infancy-onset recurrent metabolic crises with encephalocardiomyopathy. Am J Hum Genet. 2016;98(2):358–62.CrossRefPubMedPubMedCentral
335.
Zurück zum Zitat Lalani SR, Liu P, Rosenfeld JA, Watkin LB, Chiang T, Leduc MS, et al. Recurrent muscle weakness with rhabdomyolysis, metabolic crises, and cardiac arrhythmia due to bi-allelic TANGO2 mutations. Am J Hum Genet. 2016;98(2):347–57.CrossRefPubMedPubMedCentral Lalani SR, Liu P, Rosenfeld JA, Watkin LB, Chiang T, Leduc MS, et al. Recurrent muscle weakness with rhabdomyolysis, metabolic crises, and cardiac arrhythmia due to bi-allelic TANGO2 mutations. Am J Hum Genet. 2016;98(2):347–57.CrossRefPubMedPubMedCentral
336.
Zurück zum Zitat Fukata Y, Fukata M. Protein palmitoylation in neuronal development and synaptic plasticity. Nat Rev Neurosci. 2010;11(3):161–75.CrossRefPubMed Fukata Y, Fukata M. Protein palmitoylation in neuronal development and synaptic plasticity. Nat Rev Neurosci. 2010;11(3):161–75.CrossRefPubMed
337.
Zurück zum Zitat Mukai J, Dhilla A, Drew LJ, Stark KL, Cao L, MacDermott AB, et al. Palmitoylation-dependent neuro- developmental deficits in a mouse model of 22q11 microdeletion. Nat Neurosci. 2008;11(11):1302–10.CrossRefPubMedPubMedCentral Mukai J, Dhilla A, Drew LJ, Stark KL, Cao L, MacDermott AB, et al. Palmitoylation-dependent neuro- developmental deficits in a mouse model of 22q11 microdeletion. Nat Neurosci. 2008;11(11):1302–10.CrossRefPubMedPubMedCentral
338.
Zurück zum Zitat Mukai J, Tamura M, Fenelon K, Rosen AM, Spellman TJ, Kang R, et al. Molecular substrates of altered axonal growth and brain connectivity in a mouse model of schizophrenia. Neuron. 2015;86(3):680–95.CrossRefPubMedPubMedCentral Mukai J, Tamura M, Fenelon K, Rosen AM, Spellman TJ, Kang R, et al. Molecular substrates of altered axonal growth and brain connectivity in a mouse model of schizophrenia. Neuron. 2015;86(3):680–95.CrossRefPubMedPubMedCentral
339.
Zurück zum Zitat Charych EI, Akum BF, Goldberg JS, Jornsten RJ, Rongo C, Zheng JQ, et al. Activity-independent regulation of dendrite patterning by postsynaptic density protein PSD-95. J Neurosci. 2006;26(40):10164–76.CrossRefPubMedPubMedCentral Charych EI, Akum BF, Goldberg JS, Jornsten RJ, Rongo C, Zheng JQ, et al. Activity-independent regulation of dendrite patterning by postsynaptic density protein PSD-95. J Neurosci. 2006;26(40):10164–76.CrossRefPubMedPubMedCentral
340.
Zurück zum Zitat Ehrlich I, Malinow R. Postsynaptic density 95 controls AMPA receptor incorporation during long-term potentiation and experience-driven synaptic plasticity. J Neurosci. 2004;24(4):916–27.CrossRefPubMedPubMedCentral Ehrlich I, Malinow R. Postsynaptic density 95 controls AMPA receptor incorporation during long-term potentiation and experience-driven synaptic plasticity. J Neurosci. 2004;24(4):916–27.CrossRefPubMedPubMedCentral
341.
Zurück zum Zitat El-Husseini AE, Schnell E, Chetkovich DM, Nicoll RA, Bredt DS. PSD-95 involvement in maturation of excitatory synapses. Science. 2000;290(5495):1364–8.PubMed El-Husseini AE, Schnell E, Chetkovich DM, Nicoll RA, Bredt DS. PSD-95 involvement in maturation of excitatory synapses. Science. 2000;290(5495):1364–8.PubMed
342.
Zurück zum Zitat Ho GP, Selvakumar B, Mukai J, Hester LD, Wang Y, Gogos JA, et al. S-nitrosylation and S-palmitoylation reciprocally regulate synaptic targeting of PSD-95. Neuron. 2011;71(1):131–41.CrossRefPubMedPubMedCentral Ho GP, Selvakumar B, Mukai J, Hester LD, Wang Y, Gogos JA, et al. S-nitrosylation and S-palmitoylation reciprocally regulate synaptic targeting of PSD-95. Neuron. 2011;71(1):131–41.CrossRefPubMedPubMedCentral
343.
Zurück zum Zitat Thomas GM, Hayashi T, Huganir RL, Linden DJ. DHHC8-dependent PICK1 palmitoylation is required for induction of cerebellar long-term synaptic depression. J Neurosci. 2013;33(39):15401–7.CrossRefPubMedPubMedCentral Thomas GM, Hayashi T, Huganir RL, Linden DJ. DHHC8-dependent PICK1 palmitoylation is required for induction of cerebellar long-term synaptic depression. J Neurosci. 2013;33(39):15401–7.CrossRefPubMedPubMedCentral
344.
Zurück zum Zitat Ebersole B, Petko J, Woll M, Murakami S, Sokolina K, Wong V, et al. Effect of C-terminal S-palmitoylation on D2 dopamine receptor trafficking and stability. PLoS One. 2015;10(11):e0140661.CrossRefPubMedPubMedCentral Ebersole B, Petko J, Woll M, Murakami S, Sokolina K, Wong V, et al. Effect of C-terminal S-palmitoylation on D2 dopamine receptor trafficking and stability. PLoS One. 2015;10(11):e0140661.CrossRefPubMedPubMedCentral
345.
Zurück zum Zitat Mukai J, Liu H, Burt RA, Swor DE, Lai WS, Karayiorgou M, et al. Evidence that the gene encoding ZDHHC8 contributes to the risk of schizophrenia. Nat Genet. 2004;36(7):725–31.CrossRefPubMed Mukai J, Liu H, Burt RA, Swor DE, Lai WS, Karayiorgou M, et al. Evidence that the gene encoding ZDHHC8 contributes to the risk of schizophrenia. Nat Genet. 2004;36(7):725–31.CrossRefPubMed
346.
Zurück zum Zitat Giudetti AM, Stanca E, Siculella L, Gnoni GV, Damiano F. Nutritional and hormonal regulation of citrate and carnitine/acylcarnitine transporters: two mitochondrial carriers involved in fatty acid metabolism. Int J Mol Sci. 2016;17(6):817.CrossRefPubMedCentral Giudetti AM, Stanca E, Siculella L, Gnoni GV, Damiano F. Nutritional and hormonal regulation of citrate and carnitine/acylcarnitine transporters: two mitochondrial carriers involved in fatty acid metabolism. Int J Mol Sci. 2016;17(6):817.CrossRefPubMedCentral
347.
Zurück zum Zitat Morciano P, Carrisi C, Capobianco L, Mannini L, Burgio G, Cestra G, et al. A conserved role for the mitochondrial citrate transporter sea/SLC25A1 in the maintenance of chromosome integrity. Hum Mol Genet. 2009;18(21):4180–8.CrossRefPubMed Morciano P, Carrisi C, Capobianco L, Mannini L, Burgio G, Cestra G, et al. A conserved role for the mitochondrial citrate transporter sea/SLC25A1 in the maintenance of chromosome integrity. Hum Mol Genet. 2009;18(21):4180–8.CrossRefPubMed
348.
Zurück zum Zitat Catalina-Rodriguez O, Kolukula VK, Tomita Y, Preet A, Palmieri F, Wellstein A, et al. The mitochondrial citrate transporter, CIC, is essential for mitochondrial homeostasis. Oncotarget. 2012;3(10):1220–35.CrossRefPubMedPubMedCentral Catalina-Rodriguez O, Kolukula VK, Tomita Y, Preet A, Palmieri F, Wellstein A, et al. The mitochondrial citrate transporter, CIC, is essential for mitochondrial homeostasis. Oncotarget. 2012;3(10):1220–35.CrossRefPubMedPubMedCentral
349.
Zurück zum Zitat Infantino V, Iacobazzi V, Menga A, Avantaggiati ML, Palmieri F. A key role of the mitochondrial citrate carrier (SLC25A1) in TNFalpha- and IFNgamma-triggered inflammation. Biochim Biophys Acta. 2014;1839(11):1217–25.CrossRefPubMedPubMedCentral Infantino V, Iacobazzi V, Menga A, Avantaggiati ML, Palmieri F. A key role of the mitochondrial citrate carrier (SLC25A1) in TNFalpha- and IFNgamma-triggered inflammation. Biochim Biophys Acta. 2014;1839(11):1217–25.CrossRefPubMedPubMedCentral
350.
Zurück zum Zitat Majd H, King MS, Smith AC, Kunji ERS. Pathogenic mutations of the human mitochondrial citrate carrier SLC25A1 lead to impaired citrate export required for lipid, dolichol, ubiquinone and sterol synthesis. Biochim Biophys Acta Bioenerg. 2018;1859(1):1–7.CrossRefPubMed Majd H, King MS, Smith AC, Kunji ERS. Pathogenic mutations of the human mitochondrial citrate carrier SLC25A1 lead to impaired citrate export required for lipid, dolichol, ubiquinone and sterol synthesis. Biochim Biophys Acta Bioenerg. 2018;1859(1):1–7.CrossRefPubMed
351.
Zurück zum Zitat Chaouch A, Porcelli V, Cox D, Edvardson S, Scarcia P, De Grassi A, et al. Mutations in the mitochondrial citrate carrier SLC25A1 are associated with impaired neuromuscular transmission. J Neuromuscul Dis. 2014;1(1):75–90.PubMedPubMedCentral Chaouch A, Porcelli V, Cox D, Edvardson S, Scarcia P, De Grassi A, et al. Mutations in the mitochondrial citrate carrier SLC25A1 are associated with impaired neuromuscular transmission. J Neuromuscul Dis. 2014;1(1):75–90.PubMedPubMedCentral
352.
Zurück zum Zitat Nota B, Struys EA, Pop A, Jansen EE, Fernandez Ojeda MR, Kanhai WA, et al. Deficiency in SLC25A1, encoding the mitochondrial citrate carrier, causes combined D-2- and L-2-hydroxyglutaric aciduria. Am J Hum Genet. 2013;92(4):627–31.CrossRefPubMedPubMedCentral Nota B, Struys EA, Pop A, Jansen EE, Fernandez Ojeda MR, Kanhai WA, et al. Deficiency in SLC25A1, encoding the mitochondrial citrate carrier, causes combined D-2- and L-2-hydroxyglutaric aciduria. Am J Hum Genet. 2013;92(4):627–31.CrossRefPubMedPubMedCentral
353.
Zurück zum Zitat Smith A, McBride S, Marcadier JL, Michaud J, Al-Dirbashi OY, Schwartzentruber J, et al. Severe neonatal presentation of mitochondrial citrateccarrier (SLC25A1) deficiency. In: JIMD Rep 2016 Jun 16; 2016. Smith A, McBride S, Marcadier JL, Michaud J, Al-Dirbashi OY, Schwartzentruber J, et al. Severe neonatal presentation of mitochondrial citrateccarrier (SLC25A1) deficiency. In: JIMD Rep 2016 Jun 16; 2016.
354.
Zurück zum Zitat Edvardson S, Porcelli V, Jalas C, Soiferman D, Kellner Y, Shaag A, et al. Agenesis of corpus callosum and optic nerve hypoplasia due to mutations in SLC25A1 encoding the mitochondrial citrate transporter. J Med Genet. 2013;50(4):240–5.CrossRefPubMed Edvardson S, Porcelli V, Jalas C, Soiferman D, Kellner Y, Shaag A, et al. Agenesis of corpus callosum and optic nerve hypoplasia due to mutations in SLC25A1 encoding the mitochondrial citrate transporter. J Med Genet. 2013;50(4):240–5.CrossRefPubMed
355.
Zurück zum Zitat Prasun P, Young S, Salomons G, Werneke A, Jiang YH, Struys E, et al. Expanding the clinical spectrum of mitochondrial citrate carrier (SLC25A1) deficiency: facial dysmorphism in siblings with epileptic encephalopathy and combined D,L-2-hydroxyglutaric aciduria. JIMD Rep. 2015;19:111–5.PubMedPubMedCentral Prasun P, Young S, Salomons G, Werneke A, Jiang YH, Struys E, et al. Expanding the clinical spectrum of mitochondrial citrate carrier (SLC25A1) deficiency: facial dysmorphism in siblings with epileptic encephalopathy and combined D,L-2-hydroxyglutaric aciduria. JIMD Rep. 2015;19:111–5.PubMedPubMedCentral
356.
Zurück zum Zitat Eguchi M, Ozaki E, Yamauchi T, Ohta M, Higaki T, Masuda K, et al. Manifestation of recessive combined D-2-, L-2-hydroxyglutaric aciduria in combination with 22q11.2 deletion syndrome. Am J Med Genet A. 2018;176(2):351–8.CrossRefPubMed Eguchi M, Ozaki E, Yamauchi T, Ohta M, Higaki T, Masuda K, et al. Manifestation of recessive combined D-2-, L-2-hydroxyglutaric aciduria in combination with 22q11.2 deletion syndrome. Am J Med Genet A. 2018;176(2):351–8.CrossRefPubMed
357.
Zurück zum Zitat Damdimopoulos AE, Miranda-Vizuete A, Pelto-Huikko M, Gustafsson JA, Spyrou G. Human mitochondrial thioredoxin. Involvement in mitochondrial membrane potential and cell death. J Biol Chem. 2002;277(36):33249–57.CrossRefPubMed Damdimopoulos AE, Miranda-Vizuete A, Pelto-Huikko M, Gustafsson JA, Spyrou G. Human mitochondrial thioredoxin. Involvement in mitochondrial membrane potential and cell death. J Biol Chem. 2002;277(36):33249–57.CrossRefPubMed
358.
Zurück zum Zitat Turanov AA, Su D, Gladyshev VN. Characterization of alternative cytosolic forms and cellular targets of mouse mitochondrial thioredoxin reductase. J Biol Chem. 2006;281(32):22953–63.CrossRefPubMed Turanov AA, Su D, Gladyshev VN. Characterization of alternative cytosolic forms and cellular targets of mouse mitochondrial thioredoxin reductase. J Biol Chem. 2006;281(32):22953–63.CrossRefPubMed
359.
Zurück zum Zitat Conrad M. Transgenic mouse models for the vital selenoenzymes cytosolic thioredoxin reductase, mitochondrial thioredoxin reductase and glutathione peroxidase 4. Biochim Biophys Acta. 2009;1790(11):1575–85.CrossRefPubMed Conrad M. Transgenic mouse models for the vital selenoenzymes cytosolic thioredoxin reductase, mitochondrial thioredoxin reductase and glutathione peroxidase 4. Biochim Biophys Acta. 2009;1790(11):1575–85.CrossRefPubMed
360.
Zurück zum Zitat Kim MR, Chang HS, Kim BH, Kim S, Baek SH, Kim JH, et al. Involvements of mitochondrial thioredoxin reductase (TrxR2) in cell proliferation. Biochem Biophys Res Commun. 2003;304(1):119–24.CrossRefPubMed Kim MR, Chang HS, Kim BH, Kim S, Baek SH, Kim JH, et al. Involvements of mitochondrial thioredoxin reductase (TrxR2) in cell proliferation. Biochem Biophys Res Commun. 2003;304(1):119–24.CrossRefPubMed
361.
Zurück zum Zitat Tanaka T, Hosoi F, Yamaguchi-Iwai Y, Nakamura H, Masutani H, Ueda S, et al. Thioredoxin-2 (TRX-2) is an essential gene regulating mitochondria-dependent apoptosis. EMBO J. 2002;21(7):1695–703.CrossRefPubMedPubMedCentral Tanaka T, Hosoi F, Yamaguchi-Iwai Y, Nakamura H, Masutani H, Ueda S, et al. Thioredoxin-2 (TRX-2) is an essential gene regulating mitochondria-dependent apoptosis. EMBO J. 2002;21(7):1695–703.CrossRefPubMedPubMedCentral
362.
Zurück zum Zitat Conrad M, Jakupoglu C, Moreno SG, Lippl S, Banjac A, Schneider M, et al. Essential role for mitochondrial thioredoxin reductase in hematopoiesis, heart development, and heart function. Mol Cell Biol. 2004;24(21):9414–23.CrossRefPubMedPubMedCentral Conrad M, Jakupoglu C, Moreno SG, Lippl S, Banjac A, Schneider M, et al. Essential role for mitochondrial thioredoxin reductase in hematopoiesis, heart development, and heart function. Mol Cell Biol. 2004;24(21):9414–23.CrossRefPubMedPubMedCentral
363.
Zurück zum Zitat Sibbing D, Pfeufer A, Perisic T, Mannes AM, Fritz-Wolf K, Unwin S, et al. Mutations in the mitochondrial thioredoxin reductase gene TXNRD2 cause dilated cardiomyopathy. Eur Heart J. 2011;32(9):1121–33.CrossRefPubMed Sibbing D, Pfeufer A, Perisic T, Mannes AM, Fritz-Wolf K, Unwin S, et al. Mutations in the mitochondrial thioredoxin reductase gene TXNRD2 cause dilated cardiomyopathy. Eur Heart J. 2011;32(9):1121–33.CrossRefPubMed
364.
Zurück zum Zitat Horstkotte J, Perisic T, Schneider M, Lange P, Schroeder M, Kiermayer C, et al. Mitochondrial thioredoxin reductase is essential for early postischemic myocardial protection. Circulation. 2011;124(25):2892–902.CrossRefPubMed Horstkotte J, Perisic T, Schneider M, Lange P, Schroeder M, Kiermayer C, et al. Mitochondrial thioredoxin reductase is essential for early postischemic myocardial protection. Circulation. 2011;124(25):2892–902.CrossRefPubMed
365.
Zurück zum Zitat Kiermayer C, Northrup E, Schrewe A, Walch A, de Angelis MH, Schoensiegel F, et al. Heart-specific knockout of the mitochondrial thioredoxin reductase (Txnrd2) induces metabolic and contractile dysfunction in the aging myocardium. J Am Heart Assoc. 2015;4(7):e002153. Kiermayer C, Northrup E, Schrewe A, Walch A, de Angelis MH, Schoensiegel F, et al. Heart-specific knockout of the mitochondrial thioredoxin reductase (Txnrd2) induces metabolic and contractile dysfunction in the aging myocardium. J Am Heart Assoc. 2015;4(7):e002153.
366.
Zurück zum Zitat Soerensen J, Jakupoglu C, Beck H, Forster H, Schmidt J, Schmahl W, et al. The role of thioredoxin reductases in brain development. PLoS One. 2008;3(3):e1813.CrossRefPubMedPubMedCentral Soerensen J, Jakupoglu C, Beck H, Forster H, Schmidt J, Schmahl W, et al. The role of thioredoxin reductases in brain development. PLoS One. 2008;3(3):e1813.CrossRefPubMedPubMedCentral
367.
Zurück zum Zitat Beck MA, Levander OA, Handy J. Selenium deficiency and viral infection. J Nutr. 2003;133(5 Suppl 1):1463S–7S.CrossRefPubMed Beck MA, Levander OA, Handy J. Selenium deficiency and viral infection. J Nutr. 2003;133(5 Suppl 1):1463S–7S.CrossRefPubMed
368.
Zurück zum Zitat Khoso PA, Yang Z, Liu C, Li S. Selenium deficiency downregulates selenoproteins and suppresses immune function in chicken thymus. Biol Trace Elem Res. 2015;167(1):48–55.CrossRefPubMed Khoso PA, Yang Z, Liu C, Li S. Selenium deficiency downregulates selenoproteins and suppresses immune function in chicken thymus. Biol Trace Elem Res. 2015;167(1):48–55.CrossRefPubMed
369.
Zurück zum Zitat Geisberger R, Kiermayer C, Homig C, Conrad M, Schmidt J, Zimber-Strobl U, et al. B- and T-cell-specific inactivation of thioredoxin reductase 2 does not impair lymphocyte development and maintenance. Biol Chem. 2007;388(10):1083–90.CrossRefPubMed Geisberger R, Kiermayer C, Homig C, Conrad M, Schmidt J, Zimber-Strobl U, et al. B- and T-cell-specific inactivation of thioredoxin reductase 2 does not impair lymphocyte development and maintenance. Biol Chem. 2007;388(10):1083–90.CrossRefPubMed
370.
Zurück zum Zitat Ge KY, Xue A, Bai J, Wang SQ. Keshan disease - an endemic cardiomyopathy in China. Virchows Arch A Pathol Anat Histol. 1983;401(1):1–15.CrossRef Ge KY, Xue A, Bai J, Wang SQ. Keshan disease - an endemic cardiomyopathy in China. Virchows Arch A Pathol Anat Histol. 1983;401(1):1–15.CrossRef
371.
Zurück zum Zitat Andrews RK, Berndt MC. Bernard-Soulier syndrome: an update. Semin Thromb Hemost. 2013;39(6):656–62.CrossRefPubMed Andrews RK, Berndt MC. Bernard-Soulier syndrome: an update. Semin Thromb Hemost. 2013;39(6):656–62.CrossRefPubMed
372.
Zurück zum Zitat Sumitha E, Jayandharan GR, David S, Jacob RR, Sankari Devi G, Bargavi B, et al. Molecular basis of Bernard-Soulier syndrome in 27 patients from India. J Thromb Haemost. 2011;9(8):1590–8.CrossRefPubMed Sumitha E, Jayandharan GR, David S, Jacob RR, Sankari Devi G, Bargavi B, et al. Molecular basis of Bernard-Soulier syndrome in 27 patients from India. J Thromb Haemost. 2011;9(8):1590–8.CrossRefPubMed
374.
Zurück zum Zitat Rein CM, Desai UR, Church FC. Serpin-glycosaminoglycan interactions. Methods Enzymol. 2011;501:105–37.CrossRefPubMed Rein CM, Desai UR, Church FC. Serpin-glycosaminoglycan interactions. Methods Enzymol. 2011;501:105–37.CrossRefPubMed
375.
Zurück zum Zitat Aihara K, Azuma H, Akaike M, Ikeda Y, Sata M, Takamori N, et al. Strain-dependent embryonic lethality and exaggerated vascular remodeling in heparin cofactor II-deficient mice. J Clin Invest. 2007;117(6):1514–26.CrossRefPubMedPubMedCentral Aihara K, Azuma H, Akaike M, Ikeda Y, Sata M, Takamori N, et al. Strain-dependent embryonic lethality and exaggerated vascular remodeling in heparin cofactor II-deficient mice. J Clin Invest. 2007;117(6):1514–26.CrossRefPubMedPubMedCentral
376.
Zurück zum Zitat Ikeda Y, Aihara K, Yoshida S, Iwase T, Tajima S, Izawa-Ishizawa Y, et al. Heparin cofactor II, a serine protease inhibitor, promotes angiogenesis via activation of the AMP-activated protein kinase-endothelial nitric-oxide synthase signaling pathway. J Biol Chem. 2012;287(41):34256–63.CrossRefPubMedPubMedCentral Ikeda Y, Aihara K, Yoshida S, Iwase T, Tajima S, Izawa-Ishizawa Y, et al. Heparin cofactor II, a serine protease inhibitor, promotes angiogenesis via activation of the AMP-activated protein kinase-endothelial nitric-oxide synthase signaling pathway. J Biol Chem. 2012;287(41):34256–63.CrossRefPubMedPubMedCentral
377.
Zurück zum Zitat Hoogendoorn B, Coleman SL, Guy CA, Smith SK, O'Donovan MC, Buckland PR. Functional analysis of polymorphisms in the promoter regions of genes on 22q11. Hum Mutat. 2004;24(1):35–42.CrossRefPubMed Hoogendoorn B, Coleman SL, Guy CA, Smith SK, O'Donovan MC, Buckland PR. Functional analysis of polymorphisms in the promoter regions of genes on 22q11. Hum Mutat. 2004;24(1):35–42.CrossRefPubMed
378.
Zurück zum Zitat Akar NA, Adekile AD. Chromosome 22q11.2 deletion presenting with immune-mediated cytopenias, macrothrombocytopenia and platelet dysfunction. Med Princ Pract. 2007;16(4):318–20.CrossRefPubMed Akar NA, Adekile AD. Chromosome 22q11.2 deletion presenting with immune-mediated cytopenias, macrothrombocytopenia and platelet dysfunction. Med Princ Pract. 2007;16(4):318–20.CrossRefPubMed
379.
Zurück zum Zitat Rosa RF, Rosa RC, Dos Santos PP, Zen PR, Paskulin GA. Hematological abnormalities and 22q11.2 deletion syndrome. Rev Bras Hematol Hemoter. 2011;33(2):151–4.CrossRefPubMedPubMedCentral Rosa RF, Rosa RC, Dos Santos PP, Zen PR, Paskulin GA. Hematological abnormalities and 22q11.2 deletion syndrome. Rev Bras Hematol Hemoter. 2011;33(2):151–4.CrossRefPubMedPubMedCentral
380.
Zurück zum Zitat Brenner MK, Clarke S, Mahnke DK, Simpson P, Bercovitz RS, Tomita-Mitchell A, et al. Effect of 22q11.2 deletion on bleeding and transfusion utilization in children with congenital heart disease undergoing cardiac surgery. Pediatr Res. 2016;79(2):318–24.CrossRefPubMed Brenner MK, Clarke S, Mahnke DK, Simpson P, Bercovitz RS, Tomita-Mitchell A, et al. Effect of 22q11.2 deletion on bleeding and transfusion utilization in children with congenital heart disease undergoing cardiac surgery. Pediatr Res. 2016;79(2):318–24.CrossRefPubMed
381.
Zurück zum Zitat Zwifelhofer NMJ, Bercovitz RS, Weik LA, Moroi A, LaRose S, Newman PJ, et al. Hemizygosity for the gene encoding glycoprotein Ib beta (GPIbbeta) is not responsible for macrothrombocytopenia and bleeding in patients with 22q11 deletion syndrome. J Thromb Haemost. 2019;17(2):295–305.CrossRefPubMed Zwifelhofer NMJ, Bercovitz RS, Weik LA, Moroi A, LaRose S, Newman PJ, et al. Hemizygosity for the gene encoding glycoprotein Ib beta (GPIbbeta) is not responsible for macrothrombocytopenia and bleeding in patients with 22q11 deletion syndrome. J Thromb Haemost. 2019;17(2):295–305.CrossRefPubMed
382.
Zurück zum Zitat Edelmann L, Stankiewicz P, Spiteri E, Pandita RK, Shaffer L, Lupski JR, et al. Two functional copies of the DGCR6 gene are present on human chromosome 22q11 due to a duplication of an ancestral locus. Genome Res. 2001;11(2):208–17.CrossRefPubMedPubMedCentral Edelmann L, Stankiewicz P, Spiteri E, Pandita RK, Shaffer L, Lupski JR, et al. Two functional copies of the DGCR6 gene are present on human chromosome 22q11 due to a duplication of an ancestral locus. Genome Res. 2001;11(2):208–17.CrossRefPubMedPubMedCentral
383.
Zurück zum Zitat Demczuk S, Thomas G, Aurias A. Isolation of a novel gene from the DiGeorge syndrome critical region with homology to Drosophila gdl and to human LAMC1 genes. Hum Mol Genet. 1996;5(5):633–8.CrossRefPubMed Demczuk S, Thomas G, Aurias A. Isolation of a novel gene from the DiGeorge syndrome critical region with homology to Drosophila gdl and to human LAMC1 genes. Hum Mol Genet. 1996;5(5):633–8.CrossRefPubMed
384.
Zurück zum Zitat Hierck BP, Molin DG, Boot MJ, Poelmann RE, Gittenberger-de Groot AC. A chicken model for DGCR6 as a modifier gene in the DiGeorge critical region. Pediatr Res. 2004;56(3):440–8.CrossRefPubMed Hierck BP, Molin DG, Boot MJ, Poelmann RE, Gittenberger-de Groot AC. A chicken model for DGCR6 as a modifier gene in the DiGeorge critical region. Pediatr Res. 2004;56(3):440–8.CrossRefPubMed
385.
Zurück zum Zitat Lindsay EA, Baldini A. A mouse gene (Dgcr6) related to the Drosophila gonadal gene is expressed in early embryogenesis and is the homolog of a human gene deleted in DiGeorge syndrome. Cytogenet Cell Genet. 1997;79(3–4):243–7.CrossRefPubMed Lindsay EA, Baldini A. A mouse gene (Dgcr6) related to the Drosophila gonadal gene is expressed in early embryogenesis and is the homolog of a human gene deleted in DiGeorge syndrome. Cytogenet Cell Genet. 1997;79(3–4):243–7.CrossRefPubMed
386.
Zurück zum Zitat Gao W, Higaki T, Eguchi-Ishimae M, Iwabuki H, Wu Z, Yamamoto E, et al. DGCR6 at the proximal part of the DiGeorge critical region is involved in conotruncal heart defects. Hum Genome Var. 2015;2:15004.CrossRefPubMedPubMedCentral Gao W, Higaki T, Eguchi-Ishimae M, Iwabuki H, Wu Z, Yamamoto E, et al. DGCR6 at the proximal part of the DiGeorge critical region is involved in conotruncal heart defects. Hum Genome Var. 2015;2:15004.CrossRefPubMedPubMedCentral
387.
Zurück zum Zitat Das Chakraborty R, Bernal AJ, Schoch K, Howard TD, Ip EH, Hooper SR, et al. Dysregulation of DGCR6 and DGCR6L: psychopathological outcomes in chromosome 22q11.2 deletion syndrome. Transl. Psychiatry. 2012;2:e105. Das Chakraborty R, Bernal AJ, Schoch K, Howard TD, Ip EH, Hooper SR, et al. Dysregulation of DGCR6 and DGCR6L: psychopathological outcomes in chromosome 22q11.2 deletion syndrome. Transl. Psychiatry. 2012;2:e105.
388.
Zurück zum Zitat Bruderer RM, Brasseur C, Meyer HH. The AAA ATPase p97/VCP interacts with its alternative co-factors, Ufd1-Npl4 and p47, through a common bipartite binding mechanism. J Biol Chem. 2004;279(48):49609–16.CrossRefPubMed Bruderer RM, Brasseur C, Meyer HH. The AAA ATPase p97/VCP interacts with its alternative co-factors, Ufd1-Npl4 and p47, through a common bipartite binding mechanism. J Biol Chem. 2004;279(48):49609–16.CrossRefPubMed
389.
Zurück zum Zitat Bays NW, Hampton RY. Cdc48–Ufd1–Npl4: stuck in the middle with Ub. Curr biol. 2002;12(10):R366–71.CrossRefPubMed Bays NW, Hampton RY. Cdc48–Ufd1–Npl4: stuck in the middle with Ub. Curr biol. 2002;12(10):R366–71.CrossRefPubMed
390.
Zurück zum Zitat Alzayady KJ, panning MM, Kelley GG, Wojcikiewicz RJ. Involvement of the p97-Ufd1-Npl4 complex in the regulated endoplasmic reticulum-associated degradation of inositol 1,4,5-trisphosphate receptors. J Biol Chem. 2005;280(41):34530–7.CrossRefPubMed Alzayady KJ, panning MM, Kelley GG, Wojcikiewicz RJ. Involvement of the p97-Ufd1-Npl4 complex in the regulated endoplasmic reticulum-associated degradation of inositol 1,4,5-trisphosphate receptors. J Biol Chem. 2005;280(41):34530–7.CrossRefPubMed
391.
Zurück zum Zitat Kim NC, Tresse E, Kolaitis RM, Molliex A, Thomas RE, Alami NH, et al. VCP is essential for mitochondrial quality control by PINK1/Parkin and this function is impaired by VCP mutations. Neuron. 2013;78(1):65–80.CrossRefPubMedPubMedCentral Kim NC, Tresse E, Kolaitis RM, Molliex A, Thomas RE, Alami NH, et al. VCP is essential for mitochondrial quality control by PINK1/Parkin and this function is impaired by VCP mutations. Neuron. 2013;78(1):65–80.CrossRefPubMedPubMedCentral
392.
Zurück zum Zitat Kimura Y, Fukushi J, Hori S, Matsuda N, Okatsu K, Kakiyama Y, et al. Different dynamic movements of wild-type and pathogenic VCPs and their cofactors to damaged mitochondria in a Parkin-mediated mitochondrial quality control system. Genes Cells. 2013;18(12):1131–43.CrossRefPubMed Kimura Y, Fukushi J, Hori S, Matsuda N, Okatsu K, Kakiyama Y, et al. Different dynamic movements of wild-type and pathogenic VCPs and their cofactors to damaged mitochondria in a Parkin-mediated mitochondrial quality control system. Genes Cells. 2013;18(12):1131–43.CrossRefPubMed
393.
Zurück zum Zitat Cao K, Nakajima R, Meyer HH, Zheng Y. The AAA-ATPase Cdc48/p97 regulates spindle disassembly at the end of mitosis. Cell. 2003;115(3):355–67.CrossRefPubMed Cao K, Nakajima R, Meyer HH, Zheng Y. The AAA-ATPase Cdc48/p97 regulates spindle disassembly at the end of mitosis. Cell. 2003;115(3):355–67.CrossRefPubMed
394.
Zurück zum Zitat Lee JJ, Park JK, Jeong J, Jeon H, Yoon JB, Kim EE, et al. Complex of Fas-associated factor 1 (FAF1) with valosin-containing protein (VCP)-Npl4-Ufd1 and polyubiquitinated proteins promotes endoplasmic reticulum-associated degradation (ERAD). J Biol Chem. 2013;288(10):6998–7011.CrossRefPubMedPubMedCentral Lee JJ, Park JK, Jeong J, Jeon H, Yoon JB, Kim EE, et al. Complex of Fas-associated factor 1 (FAF1) with valosin-containing protein (VCP)-Npl4-Ufd1 and polyubiquitinated proteins promotes endoplasmic reticulum-associated degradation (ERAD). J Biol Chem. 2013;288(10):6998–7011.CrossRefPubMedPubMedCentral
395.
Zurück zum Zitat Lin KW, McDonald KR, Guise AJ, Chan A, Cristea IM, Zakian VA. Proteomics of yeast telomerase identified Cdc48-Npl4-Ufd1 and Ufd4 as regulators of Est1 and telomere length. Nat Commun. 2015;6:8290.CrossRefPubMed Lin KW, McDonald KR, Guise AJ, Chan A, Cristea IM, Zakian VA. Proteomics of yeast telomerase identified Cdc48-Npl4-Ufd1 and Ufd4 as regulators of Est1 and telomere length. Nat Commun. 2015;6:8290.CrossRefPubMed
396.
Zurück zum Zitat Riemer A, Dobrynin G, Dressler A, Bremer S, Soni A, Iliakis G, et al. The p97-Ufd1-Npl4 ATPase complex ensures robustness of the G2/M checkpoint by facilitating CDC25A degradation. Cell Cycle. 2014;13(6):919–27.CrossRefPubMedPubMedCentral Riemer A, Dobrynin G, Dressler A, Bremer S, Soni A, Iliakis G, et al. The p97-Ufd1-Npl4 ATPase complex ensures robustness of the G2/M checkpoint by facilitating CDC25A degradation. Cell Cycle. 2014;13(6):919–27.CrossRefPubMedPubMedCentral
397.
Zurück zum Zitat Verma R, Oania RS, Kolawa NJ, Deshaies RJ. Cdc48/p97 promotes degradation of aberrant nascent polypeptides bound to the ribosome. Elife. 2013;2:e00308.CrossRefPubMedPubMedCentral Verma R, Oania RS, Kolawa NJ, Deshaies RJ. Cdc48/p97 promotes degradation of aberrant nascent polypeptides bound to the ribosome. Elife. 2013;2:e00308.CrossRefPubMedPubMedCentral
398.
Zurück zum Zitat Yamagishi C, Hierck BP, Gittenberger-De Groot AC, Yamagishi H, Srivastava D. Functional attenuation of UFD1l, a 22q11.2 deletion syndrome candidate gene, leads to cardiac outflow septation defects in chicken embryos. Pediatr Res. 2003;53(4):546–53.CrossRefPubMed Yamagishi C, Hierck BP, Gittenberger-De Groot AC, Yamagishi H, Srivastava D. Functional attenuation of UFD1l, a 22q11.2 deletion syndrome candidate gene, leads to cardiac outflow septation defects in chicken embryos. Pediatr Res. 2003;53(4):546–53.CrossRefPubMed
399.
Zurück zum Zitat Yamagishi H, Garg V, Matsuoka R, Thomas T, Srivastava D. A molecular pathway revealing a genetic basis for human cardiac and craniofacial defects. Science. 1999;283(5405):1158–61.CrossRefPubMed Yamagishi H, Garg V, Matsuoka R, Thomas T, Srivastava D. A molecular pathway revealing a genetic basis for human cardiac and craniofacial defects. Science. 1999;283(5405):1158–61.CrossRefPubMed
400.
Zurück zum Zitat Mohamed SA, Hanke T, Schlueter C, Bullerdiek J, Sievers HH. Ubiquitin fusion degradation 1-like gene dysregulation in bicuspid aortic valve. J Thorac Cardiovasc Surg. 2005;130(6):1531–6.CrossRefPubMed Mohamed SA, Hanke T, Schlueter C, Bullerdiek J, Sievers HH. Ubiquitin fusion degradation 1-like gene dysregulation in bicuspid aortic valve. J Thorac Cardiovasc Surg. 2005;130(6):1531–6.CrossRefPubMed
401.
Zurück zum Zitat Byrne DJ, Harmon MJ, Simpson JC, Blackstone C, O'Sullivan NC. Roles for the VCP co-factors Npl4 and Ufd1 in neuronal function in Drosophila melanogaster. J Genet Genomics. 2017;44(10):493–501.CrossRefPubMedPubMedCentral Byrne DJ, Harmon MJ, Simpson JC, Blackstone C, O'Sullivan NC. Roles for the VCP co-factors Npl4 and Ufd1 in neuronal function in Drosophila melanogaster. J Genet Genomics. 2017;44(10):493–501.CrossRefPubMedPubMedCentral
402.
Zurück zum Zitat De Luca A, Pasini A, Amati F, Botta A, Spalletta G, Alimenti S, et al. Association study of a promoter polymorphism of UFD1L gene with schizophrenia. Am J Med Genet. 2001;105(6):529–33.CrossRefPubMed De Luca A, Pasini A, Amati F, Botta A, Spalletta G, Alimenti S, et al. Association study of a promoter polymorphism of UFD1L gene with schizophrenia. Am J Med Genet. 2001;105(6):529–33.CrossRefPubMed
403.
Zurück zum Zitat Shashi V, Francis A, Hooper SR, Kranz PG, Zapadka M, Schoch K, et al. Increased corpus callosum volume in children with chromosome 22q11.2 deletion syndrome is associated with neurocognitive deficits and genetic polymorphisms. Eur J Hum Genet. 2012;20(10):1051–7.CrossRefPubMedPubMedCentral Shashi V, Francis A, Hooper SR, Kranz PG, Zapadka M, Schoch K, et al. Increased corpus callosum volume in children with chromosome 22q11.2 deletion syndrome is associated with neurocognitive deficits and genetic polymorphisms. Eur J Hum Genet. 2012;20(10):1051–7.CrossRefPubMedPubMedCentral
404.
Zurück zum Zitat Männistö PT, Kaakkola S. Catechol-O-methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors. Pharmacol Rev. 1999;5(14):593–628. Männistö PT, Kaakkola S. Catechol-O-methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors. Pharmacol Rev. 1999;5(14):593–628.
405.
Zurück zum Zitat Lundström K, Salminen M, Jalanko A, Savolainen R, Ulmanen I. Cloning and characterization of human placental catechol-O-methyltransferase cDNA. DNA Cell Biol. 1991;10(3):181–9.CrossRefPubMed Lundström K, Salminen M, Jalanko A, Savolainen R, Ulmanen I. Cloning and characterization of human placental catechol-O-methyltransferase cDNA. DNA Cell Biol. 1991;10(3):181–9.CrossRefPubMed
406.
Zurück zum Zitat Babovic D, O'Tuathaigh CM, O'Connor AM, O'Sullivan GJ, Tighe O, Croke DT, et al. Phenotypic characterization of cognition and social behavior in mice with heterozygous versus homozygous deletion of catechol-O-methyltransferase. Neuroscience. 2008;155(4):1021–9.CrossRefPubMed Babovic D, O'Tuathaigh CM, O'Connor AM, O'Sullivan GJ, Tighe O, Croke DT, et al. Phenotypic characterization of cognition and social behavior in mice with heterozygous versus homozygous deletion of catechol-O-methyltransferase. Neuroscience. 2008;155(4):1021–9.CrossRefPubMed
407.
Zurück zum Zitat Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, et al. Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci U S A. 1998;95(17):9991–6.CrossRefPubMedPubMedCentral Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, et al. Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci U S A. 1998;95(17):9991–6.CrossRefPubMedPubMedCentral
408.
409.
Zurück zum Zitat Hosak L. Role of the COMT gene Val158Met polymorphism in mental disorders: a review. Eur Psychiatry. 2007;22(5):276–81.CrossRefPubMed Hosak L. Role of the COMT gene Val158Met polymorphism in mental disorders: a review. Eur Psychiatry. 2007;22(5):276–81.CrossRefPubMed
410.
Zurück zum Zitat Wahlstrom D, White T, Hooper CJ, Vrshek-Schallhorn S, Oetting WS, Brott MJ, et al. Variations in the catechol O-methyltransferase polymorphism and prefrontally guided behaviors in adolescents. Biol Psychiatry. 2007;61(5):626–32.CrossRefPubMed Wahlstrom D, White T, Hooper CJ, Vrshek-Schallhorn S, Oetting WS, Brott MJ, et al. Variations in the catechol O-methyltransferase polymorphism and prefrontally guided behaviors in adolescents. Biol Psychiatry. 2007;61(5):626–32.CrossRefPubMed
412.
Zurück zum Zitat Hao Z, Jha KN, Kim YH, Vemuganti S, Westbrook VA, Chertihin O, et al. Expression analysis of the human testis-specific serine/threonine kinase (TSSK) homologues. A TSSK member is present in the equatorial segment of human sperm. Mol Hum Reprod. 2004;10(6):433–44.CrossRefPubMed Hao Z, Jha KN, Kim YH, Vemuganti S, Westbrook VA, Chertihin O, et al. Expression analysis of the human testis-specific serine/threonine kinase (TSSK) homologues. A TSSK member is present in the equatorial segment of human sperm. Mol Hum Reprod. 2004;10(6):433–44.CrossRefPubMed
414.
Zurück zum Zitat Towns WL, Begley TJ. Transfer RNA methytransferases and their corresponding modifications in budding yeast and humans: activities, predications, and potential roles in human health. DNA Cell Biol. 2012;31(4):434–54.CrossRefPubMedPubMedCentral Towns WL, Begley TJ. Transfer RNA methytransferases and their corresponding modifications in budding yeast and humans: activities, predications, and potential roles in human health. DNA Cell Biol. 2012;31(4):434–54.CrossRefPubMedPubMedCentral
416.
Zurück zum Zitat Hu T, Yamagishi H, Maeda J, McAnally J, Yamagishi C, Srivastava D. Tbx1 regulates fibroblast growth factors in the anterior heart field through a reinforcing autoregulatory loop involving forkhead transcription f actors. Development. 2004;131(21):5491–502.CrossRefPubMed Hu T, Yamagishi H, Maeda J, McAnally J, Yamagishi C, Srivastava D. Tbx1 regulates fibroblast growth factors in the anterior heart field through a reinforcing autoregulatory loop involving forkhead transcription f actors. Development. 2004;131(21):5491–502.CrossRefPubMed
418.
Zurück zum Zitat Maehama T, Taylor GS, Dixon JE. PTEN and myotubularin: novel phosphoinositide phosphatases. Annu Rev Biochem. 2001;70:247–79.CrossRefPubMed Maehama T, Taylor GS, Dixon JE. PTEN and myotubularin: novel phosphoinositide phosphatases. Annu Rev Biochem. 2001;70:247–79.CrossRefPubMed
Metadaten
Titel
In the line-up: deleted genes associated with DiGeorge/22q11.2 deletion syndrome: are they all suspects?
verfasst von
Zahra Motahari
Sally Ann Moody
Thomas Michael Maynard
Anthony-Samuel LaMantia
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Neurodevelopmental Disorders / Ausgabe 1/2019
Print ISSN: 1866-1947
Elektronische ISSN: 1866-1955
DOI
https://doi.org/10.1186/s11689-019-9267-z

Weitere Artikel der Ausgabe 1/2019

Journal of Neurodevelopmental Disorders 1/2019 Zur Ausgabe

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.

Weniger postpartale Depressionen nach Esketamin-Einmalgabe

Bislang gibt es kein Medikament zur Prävention von Wochenbettdepressionen. Das Injektionsanästhetikum Esketamin könnte womöglich diese Lücke füllen.

„Psychotherapie ist auch bei sehr alten Menschen hochwirksam!“

22.04.2024 DGIM 2024 Kongressbericht

Die Kombination aus Medikamenten und Psychotherapie gilt als effektivster Ansatz bei Depressionen. Das ist bei betagten Menschen nicht anders, trotz Besonderheiten.

Auf diese Krankheiten bei Geflüchteten sollten Sie vorbereitet sein

22.04.2024 DGIM 2024 Nachrichten

Um Menschen nach der Flucht aus einem Krisengebiet bestmöglich medizinisch betreuen zu können, ist es gut zu wissen, welche Erkrankungen im jeweiligen Herkunftsland häufig sind. Dabei hilft eine Internetseite der CDC (Centers for Disease Control and Prevention).