Skip to main content
Erschienen in: Journal of Clinical Immunology 2/2022

Open Access 30.10.2021 | Original Article

Biomarkers of DNA Damage Response Enable Flow Cytometry-Based Diagnostic to Identify Inborn DNA Repair Defects in Primary Immunodeficiencies

verfasst von: Kerstin Felgentreff, Ulrich Baumann, Christian Klemann, Catharina Schuetz, Dorothee Viemann, Martin Wetzke, Ulrich Pannicke, Sandra von Hardenberg, Bernd Auber, Klaus-Michael Debatin, Eva-Maria Jacobsen, Manfred Hoenig, Ansgar Schulz, Klaus Schwarz

Erschienen in: Journal of Clinical Immunology | Ausgabe 2/2022

Abstract

DNA damage is a constant event in every cell caused by exogenous factors such as ultraviolet and ionizing radiation (UVR/IR) and intercalating drugs, or endogenous metabolic and replicative stress. Proteins of the DNA damage response (DDR) network sense DNA lesions and induce cell cycle arrest, DNA repair, and apoptosis. Genetic defects of DDR or DNA repair proteins can be associated with immunodeficiency, bone marrow failure syndromes, and cancer susceptibility. Although various diagnostic tools are available to evaluate DNA damage, their quality to identify DNA repair deficiencies differs enormously and depends on affected pathways. In this study, we investigated the DDR biomarkers γH2AX (Ser139), p-ATM (Ser1981), and p-CHK2 (Thr68) using flow cytometry on peripheral blood cells obtained from patients with combined immunodeficiencies due to non-homologous end-joining (NHEJ) defects and ataxia telangiectasia (AT) in response to low-dose IR. Significantly reduced induction of all three markers was observed in AT patients compared to controls. However, delayed downregulation of γH2AX was found in patients with NHEJ defects. In contrast to previous reports of DDR in cellular models, these biomarkers were not sensitive enough to identify ARTEMIS deficiency with sufficient reliability. In summary, DDR biomarkers are suitable for diagnosing NHEJ defects and AT, which can be useful in neonates with abnormal TREC levels (T cell receptor excision circles) identified by newborn screening. We conclude that DDR biomarkers have benefits and some limitations depending on the underlying DNA repair deficiency.

Introduction

Every cell is constantly exposed to DNA damage caused by external factors such as ionizing (IR) and ultraviolet radiation (UVR), chemicals — including alkylating drugs, or by endogenous factors such as replicative and metabolic stress. While these insults may result in both DNA single-strand breaks (SSBs) and double-strand breaks (DSBs), the latter are more critical in terms of cell survival and mutation probability. Furthermore, complex lesions of base dimers and interstrand cross-links (ICL) can be induced by chemical reactions or UVR [1]. Importantly, DNA DSBs are also physiologically introduced in the T cell receptor (TCR) and immunoglobulin (Ig) genes during V(D)J recombination and class switch recombination of developing lymphocytes [2].
The cellular integrity relies on a complex network of proteins that ensure immediate sensing and efficient repair to protect the DNA from any persisting damage, known as DNA damage response (DDR). If this system fails, apoptosis, senescence, or introduction of chromosomal breaks and mutations potentially leading to neoplastic transformation are the consequences [3].
At least five DNA repair pathways — base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), non-homologous end-joining (NHEJ), and homologous recombination (HR) — are active through different stages of the cell cycle and used depending on the type of DNA lesion [1]. Inborn errors of proteins associated with DDR, DNA repair, and DNA stability result in various DNA repair deficiencies that can be associated with immunodeficiency, bone marrow failure, and cancer susceptibilities (Table 1).
Table 1
Human DNA repair deficiencies include a broad spectrum of diseases that require specific diagnostic approaches
https://static-content.springer.com/image/art%3A10.1007%2Fs10875-021-01156-7/MediaObjects/10875_2021_1156_Tab1_HTML.png
Inborn errors of DNA stability and repair may result in immunodeficiency, bone marrow failure, and susceptibility to cancer. This table provides an overview of these rare genetic diseases, their common clinical presentation, and suitable diagnostic approaches
SCID, severe combined immunodeficiency; DDR, DNA damage response; IR, ionizing radiation; MMC, mitomycin C; SCE, sister chromatid exchange; BMF, bone marrow failure; DEB, diepoxybutane; ICF, immunodeficiency, centromere instability, facial anomalies; SHM, somatic hypermutation; MSI, microsatellite instability; IF, immunofluorescence; ALPS, autoimmune lymphoproliferative syndrome
In NHEJ-mediated repair, KU70 and KU80 are the essential sensors of free DNA ends. They bind and stabilize the DNA and further recruit the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs). Together, they form an active serine/threonine DNA-PK holoenzyme that belongs to the phosphatidylinositol 3-kinase-related kinases (PIKKs) family. This complex recruits the endonuclease ARTEMIS, which processes the DNA ends with overhangs, the XRCC4/Ligase 4 heterodimer, XLF and PAXX (paralog of XRCC4 and XLF) protein to complete the repair process [4]. Genetic defects in NHEJ-associated proteins result in severe combined immunodeficiency with a lack of T and B lymphocytes and increased cellular radiation sensitivity (RS-SCID) [5]. Most of RS-SCID patients are affected by ARTEMIS deficiency caused by genomic mutations in the DCLRE1C gene and present with mild radiosensitivity [6, 7]. In contrast, DNA Ligase 4 (LIG4) deficiency results in a variable phenotypic spectrum of growth failure, microcephaly, developmental delay, and a tendency to develop bone marrow failure, myelodysplasia, and/or malignancies [810]. Deficiency of XLF causes a rare combined immunodeficiency syndrome associated with microcephaly and developmental delay [11, 12]. So far, six patients with DNA-PKcs deficiency and variable manifestations of immunodeficiency, autoimmunity, and granulomas have been identified [1316]. Of note, defects in XRCC4 [1720] and PAXX [21, 22], two factors in the LIG4-ligation complex, lead to a severe DNA repair defect, but not to an immunodeficiency and are dispensable for V(D)J recombination [23].
A major sensor of DNA DSB is the MRN complex formed by MRE11, RAD50, and NBS1 that hooks free DNA ends and activates the protein kinase ATM (ataxia telangiectasia mutated) that also belongs to the PIKK family [24]. ATM is a central player in the regulation of cell cycle checkpoints, cell survival, and DNA repair [25]. Genetic defects in ATM lead to ataxia telangiectasia (AT) presenting with cerebellar degeneration, severe cellular sensitivity to IR, genomic instability with a predisposition to cancer [26], and antibody deficiency due to impaired class switch recombination [27]. Patients with defects in proteins of the MRN complex may present with AT-like disorder (ATLD) [28, 29], Nijmegen-Breakage Syndrome (NBS) characterized by short stature, microcephaly, “bird-like” face, mental retardation, immunodeficiency, and predisposition to develop cancers [30, 31], or NBS-like syndrome [32, 33].
Defects in the HR pathway rarely result in immunodeficiency, but rather predispose to the early development of a wide variety of cancers due to chromosomal instability. Bloom syndrome (BLM) is caused by a defect in a RecQ helicase and may present with growth deficiency, a UV-sensitive skin rash, endocrine disorders, immunodeficiency, and increased susceptibility to the development of various cancers [34]. In contrast to defects of NHEJ factors with defined overlapping function in the repair of DSB caused by V(D)J recombination, immune deficiency predominantly results from impaired development and differentiation of B lymphocytes [35].
The Fanconi Anemia (FA) complex consists of at least 22 proteins, many operating in HR, such as BRCA1, BRCA2, and RAD51 [36]. FA-proteins are needed for repair of ICLs, and induction of HR to resolve DNA DSB after ICL resection. Patients with FA can present with multiple congenital abnormalities, bone marrow failure, endocrine dysfunction, and cancer [36]. Mild immunodeficiency with impaired B and NK cell function has been reported in FA patients [37].
UV radiation-induced pyrimidine dimers are recognized and excised by proteins functioning in the NER pathway. Individuals affected by NER-deficiencies are highly sensitive to sunlight that predisposes towards benign and malignant skin tumors, and can manifest neurological abnormalities including mental retardation [38]. Defects in the BER pathway, which operates in repairing small base lesions, may lead to B cell tumors and autoimmunity, since BER is involved in mutagenic pathways of B cell development during immunoglobulin class switch recombination (CSR) and somatic hypermutation (SHM) [39]. MMR acts on the same lesions as BER and corrects errors that spontaneously occur during DNA replication, including base mismatches, short insertions, or deletions. Cells with MMR-deficiency accumulate errors leading to microsatellite instability (MSI) [40]. Besides cancer susceptibility, abnormal SHM has been reported in B cells of patients affected by MMR defects [41].
Defects of DNA telomerases and methyltransferases result in dyskeratosis congenita (DC) [42], or immunodeficiency, centromeric instability, and facial anomaly syndromes (ICF) 1–4 [43], respectively, which are associated with immunodeficiency, bone marrow failure, and cancer susceptibility. Patients affected by DC may present with a broad spectrum of combined immunodeficiency, including common variable immunodeficiency (CVID) [44, 45]. The immune defect observed in ICF syndromes is associated with impaired NHEJ and B cell development [4648].
A recent diagnostic tool to approach DNA repair deficiency is to score the activation of DDR proteins that are phosphorylated in response to DNA damage. The histone protein H2AX is phosphorylated at Ser139 (γH2AX) by PIKKs ATM, ATR, and DNA-PKcs at sites of DNA DSBs forming nuclear γH2AX foci (Fig. 1A, B). After repair of DNA lesions, γH2AX is de-phosphorylated by wild-type p53-induced phosphatase 1 (WIP1) [49], protein phosphates 2A (PP2A) [50], and PP4 [51]. Kinetics analyzed at time points between 1 and 48 h after IR can be used to evaluate DNA repair capacity. Since γH2AX formation at sides of DNA DSB was discovered, it became a popular target for the assessment of DNA damage [52]. Besides immunofluorescent staining (Fig. 1B), γH2AX foci formation can also be studied by flow cytometry in suspension cells [53].
In this study, we evaluated the activation of γH2AX (Ser139), p-ATM (Ser1981), and p-CHK2 (Thr68) in PBMCs treated with IR by flow cytometry. These markers turned out to be beneficial to detect NHEJ-defects and AT with different efficiencies. We further discuss the applicability of DDR biomarkers for diagnostic purposes and alternative approaches.

Material and Methods

Patient Recruitment, PBMCs Isolation, Cell Culture

Patients diagnosed with AT, and immunodeficiency syndromes, including ARTEMIS-, XLF-, DNA-PKcs-deficiencies, as well as ICF1, ICF2, and ICF4 defects, and healthy controls were recruited from University Medical Centers of Ulm, Hannover, and Dresden (Table 2). In addition, patients affected by FA, DC, and deficiencies in the MMR (PMS2 mutation) and the SWI/SNF pathways (SMARCA4 mutation) were included, although results are not reported. All patients and controls gave informed consent, and this study was approved by the ethic committees of Ulm University (407/16), Technical University of Dresden (TUD) (BO-EK-213052020), and Hannover Medical School (MHH) (9492-BO-K-2020). Peripheral blood mononuclear cells (PBMCs) were isolated from healthy donors (HD) and patients using Ficoll Paque Plus (GE Healthcare) according to manufacturer’s instructions. PBMCs were cultured in RPMI media supplemented with 15% fetal bovine serum (FCS) (BioWhittaker™), 1% Glutamine (Thermo Fisher Scientific), 1% non-essential amino acids (Thermo Fisher Scientific), 1% penicillin/streptavidine (Thermo Fisher Scientific), and 100 U/ml hIL-2 (R&D Systems) at 1 × 10^6/ml in a 24-well culture dish (CellStar®, Greiner Bio-One) at 37 °C 5% CO2 for 96 h.
Table 2
Summary of patients included in this study
Patient ID
Gene
Protein
Clinical presentation
Age at analysis
Biomaterial (PBMC)
ART1
DCLRE1C
ARTEMIS
SCID T-B-NK + 
6 mo
Cryopreserved
ART2
DCLRE1C
ARTEMIS
SCID T-B-NK + 
6 mo
Cryopreserved
ART3
DCLRE1C
ARTEMIS
CID T + B-NK + , severe VZV, skin abscess, vitiligo
8.5 y
Fresh
ART4
DCLRE1C
ARTEMIS
CID T + B-NK + , Omenn syndrome
4 mo
Cryopreserved
ART5
DCLRE1C
ARTEMIS
SCID T-B-NK + MFT
4 mo
Cryopreserved
DNA-PKcs1
PRKDC
DNA-PKcs
CID, recurrent pneumonia, alopecia areata
5.5 y
Fresh
XLF1
NHEJ1
CERNUNNOS/XLF
CID, agammaglobulinemia, recurrent pneumonia, chronic arthritis (adenovirus type 1), microcephaly
17.5 y
Fresh
AT1
ATM
ATM
B-Zell NHL
17 y
Fresh
AT2
ATM
ATM
Recurrent pulmonary infections, ataxia
5.5 y
Fresh
AT3
ATM
ATM
Identified by newborn screening
3 mo
Fresh
AT4
ATM
ATM
Ataxia
3.5 y
Fresh
AT5
ATM
ATM
Ataxia
3.5 y
Fresh
AT6
ATM
ATM
Ataxia, granuloma
28 y
Fresh
AT7
ATM
ATM
Ataxia
4.5 y
Fresh
ICF1
DNMT3B
DNMT3B
Chronic lung disease, hepatopathy, thrombocytopenia, anemia, lymphopenia, hypogammaglobulinemia
21 y
Cryopreserved
ICF2
ZBTB24
ZBTB24
Chronic lung disease, history of CMV-pneumonia, hepatopathy, metabolic disorder, hypogammaglobulinemia
3 y
Fresh
ICF4
HELLS
HELLS
Chronic bronchitis, mental retardation, deafness, blindness, seizures, facial dysmorphia
3.5 y
Fresh
Patients affected by inborn radiosensitive immunodeficiencies such as ARTEMIS-, DNA-PKcs-, XLF-deficiency, AT, or ICF syndromes were included in this study after giving informed consent. This table summarizes basic information of affected genes, clinical presentation, age at analysis, and sample material used
SCID, severe combined immunodeficiency; CID, combined immunodeficiency; PBMC, peripheral blood mononuclear cells; VZV, Varicella zoster virus; MFT, maternal–fetal T cell transfusion; NHL, non-Hodgkin lymphoma; ICF, immunodeficiency, centromere instability, facial anomalies; CMV, cytomegalovirus

DNA Damage Induction, Fixation and Permeabilization

Cells were treated with 2 Gy of gamma radiation and fixed after 1 h, 4 h, 8 h, and 24 h using the solution A of Fix&Perm (Thermo Fisher Scientific) diluted 1:1 with phosphor buffered saline (PBS) (Thermo Fisher Scientific). After incubation at ambient temperature (RT) for 10 min, 2 ml chilled methanol (Roth) was added to each sample. Samples were stored at – 20 °C for at least 10 min up to 1 week.

Surface and Intranuclear Staining for DDR Analysis

Prior fixation, PBMCs were stained with mouse anti-human CD45 Krome-Orange (Beckman Coulter), BV421 anti-human CD3 (BD Bioscience), anti-human CD56 APC (BioLegend), anti-human CD16 APC-fire (BioLegend) 1:100 in PBS supplemented with 1% FCS, and 2 mM EDTA for 30 min at RT. After permeabilization with methanol, PBMCs were washed twice with PBS/1%FCS and stained with anti-γH2AX (Ser139) FITC (clone JBW301) (Merck Millipore) 1:250, anti-p-ATM (Ser1981) PE (BioLegend) 1:200, or anti-p-CHK2 (Thr68) PE (eBioscience, Thermo Fisher Scientific) 1:50, respectively, in PBS/1%FCS for 1 h at RT. FITC Mili-Mark™ anti-mouse IgG1-k, clone MOPC-21 (Merck Millipore), and PE anti-mouse IgG1-k (BioLegend) isotype controls were used in the same concentrations as primary antibodies. Cells were washed twice, and analyzed on Navios (Beckman Coulter), or FACSAria™ (BD) flow cytometers, respectively. PBMCs obtained from AT patients were stained with surface markers after permeabilization using anti-human CD45 APC-Cy7 (BD Bioscience), anti-human CD3 APC (BioLegend) 1:100 in PBS/1%FCS for 30 min at RT, followed by staining with intranuclear markers mentioned above.
For quality control and screening of recombination capacity, non-fixed PBMCs were stained with mouse anti-human CD3 APC-Cy7 (BioLegend), anti-human CD19 FITC (BioLegend), anti-human CD56 APC (BioLegend), anti-human CD16 PE-Cy7 (BioLegend), and anti-TCRaV7.2 (BioLegend) 1:100 in PBS/1%FCS for 20 min at RT before acquisition on a FACSAria™ (BD).

Data Analysis

Flow cytometry data were analyzed using FlowJo Vs 10.0 software. Mean fluorescent intensities (MFI) of γH2AX, p-ATM, and p-CHK2 were calculated on T and NK-cell subsets. To compare MFIs of DDR markers generated from separate experiments, fold inductions of γH2AX, p-ATM, and p-CHK2 were calculated by normalizing on MFIs of untreated samples.
Statistical analysis and generation of graphs were performed using Prism v9 software. Statistical significance was calculated by 2-way ANOVA and Tukey’s multiple comparison test. P-values ≤ 0.05 were considered significant (* p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001).

Results

DDR Quantified by H2AX Phosphorylation Can Be Used for Diagnostic Purposes in Radiosensitive Immunodeficiencies

Patients diagnosed with radiosensitive combined immunodeficiencies, such as ARTEMIS-, XLF- and DNA-PKcs-deficiencies, were recruited and studied for DDR (Table 2). PBMCs were either freshly isolated from routine blood draws or obtained from cryopreserved samples. After culture for 4 days to recover from metabolic stress, PBMCs were treated with 2 Gy of IR. The γH2AX response was investigated by flow cytometry in CD45+CD3+ T and CD45+CD3CD56dimCD16+ NK lymphocytes before IR, and 1 h, 4 h, 8 h, and 24 h after DNA damage was induced (Figure S1). Isotype controls were analyzed to define unspecific background which was subtracted from mean fluorescent intensities (MFI) of irradiated samples. Despite several limitations regarding specificity, phosphorylation of H2AX can be used as a readout for DNA DSB and the kinetics of their repair [54].
Besides V(D)J recombination in lymphocyte development, the endonuclease ARTEMIS is needed for end-processing of DNA breaks with overhangs that are repaired in a slower kinetic pathway [55]. Therefore, a delayed DNA repair process can be observed in patients with ARTEMIS deficiency 24–48 h after DNA damage has been induced [56, 57]. However, in contrast to investigations made on cell lines including human fibroblasts [58], we observed elevated levels of γH2AX in ARTEMIS patients compared to controls 24 h after DNA damage without significance (Fig. 2AD, Figure S1). Of note, T cell subsets were present in two patients affected by hypomorphic ARTEMIS deficiency (ART3 and ART4). In contrast, T and NK lymphocytes from patients with other NHEJ-DNA repair deficiencies, such as DNA-PKcs and XLF deficiency, presented with elevated γH2AX levels compared to HD in response to IR (Fig. 2E, F). This could be observed at all time points and matches previous results [59]. A side-by-side comparison of mean γH2AX MFIs obtained from all healthy controls and patients revealed significant differences at 1 h, 4 h, and 8 h after IR for patients with XLF and DNA-PKcs, but not ARTEMIS deficiency (Fig. 2G).
As reported recently, genetic variants in DNA methyltransferases or helicases, resulting in ICF syndromes, also impact on NHEJ [46, 47]. We investigated three patients affected by genetic variants in DNMT3B (ICF1), ZBTB24 (ICF2), and HELLS (ICF4) for their γH2AX-, p-ATM, and p-CHK2-mediated DDR in response to 2 Gy IR. Compared to healthy donors, increased γH2AX and p-ATM activation could be observed in patients affected by ICF1 and ICF4, however, not in the patient with ICF2 (data not shown). Very little is known about NHEJ-mediated DNA DSB repair in ICF syndromes so far. However, ICF syndromes could potentially be identified with impaired γH2AX de-phosphorylation in response to IR, and need to be differentiated from RS-(S)CIDs.
No abnormalities in DDR could be observed in PBMCs obtained from patients diagnosed with FA (FANCA, FANCC), DC (RTLE1), defects in MMR (PMS2), and the SWI/SNF complex (SMARCA4) (data not shown), although delayed reduction of γH2AX foci has been reported in FA cell lines [60].
In conclusion, γH2AX MFIs analyzed in lymphocyte subsets can be used to identify DNA repair deficiencies in the NHEJ pathway; however, the sensitivity might be too low to identify ARTEMIS defects. Although flow cytometry is faster and easier available, immunofluorescent staining of γH2AX foci in patient fibroblast lines seems to be more sensitive. Furthermore, delayed γH2AX downregulation may also be observed in patients with ICF syndromes.

DDR Analysis as a Tool to Diagnose Ataxia Telangiectasia

ATM is a key factor in the DDR and a PIK kinase that phosphorylates many downstream factors including γH2AX (Ser139), p-CHK2 (Thr68), and ATM itself (Ser1981). We investigated the activation of these three DDR biomarkers in response to low dose IR with 2 Gy in CD45+CD3+ T lymphocytes from seven patients diagnosed with AT (Fig. 3). In order to compare MFIs obtained from different experiments, fold inductions of DDR biomarkers were calculated based on basal levels measured in unirradiated samples. In contrast to NHEJ-deficient lymphocytes, upregulation of DDR markers in early response to DNA damage was severely abrogated in T cells from AT patients compared to 21 healthy controls. Whereas almost no induction of γH2AX (Fig. 3A), p-ATM (Fig. 3B), and p-CHK2 (Fig. 3C) could be observed in patients, all biomarkers were activated in the controls and levels declined over time. Therefore, differences were highly significant at early time points (1 h) after IR. In particular, p-CHK2 was identified with the best selectivity, due to the highest difference among groups of patients and controls.
In summary, DDR represented by phosphorylation of H2AX, ATM, and CHK2 is a highly sensitive tool to identify patients with AT that can be readily distinguished from patients with other combined immunodeficiencies.

Screening of V(D)J Recombination Capacity by TCRaV7.2 Expression Can Improve the Sensitivity of DDR Response

In addition to DDR, we investigated the expression of T cell receptor alpha V7.2 (TCRaV7.2) on CD45+CD3+ T cells of all patients analyzed, including an additional patient with ARTEMIS deficiency and maternal–fetal T cell transfusion (ART5), and 21 healthy controls (Fig. 4, Figure S2). TCR-Va7.2 is a TRAV segment expressed by human T lymphocytes that represents the most distal TCRa segment and has therefore been described as a marker for recombination efficacy [61]. As reported previously, no TCRaV7.2 expression was found on T cells with ARTEMIS- or DNA-PKcs-deficiency and was reduced in XLF-deficiency and on maternal T cells of ART5 and XLF1 (Fig. 4). Of note, TCRaV7.2 expression varied among AT patients, whereas most patients were characterized by lower expression compared to controls as reported previously [61]. TCRaV7.2 expression on CD3+ T cells was not altered in two patients with ICF syndromes.
Although TCRaV7.2 expression might not be as specific as sequencing of TRA or TRB genes or expression analysis of various TCRVb clones, it can be used as an additional screening marker to identify reduced recombination capacity in patients with inconclusive DDR.

Discussion

Inborn errors of DNA repair may lead to immunodeficiency, bone marrow failure, or cancer susceptibility and can be associated with growth delay, malformations, and neurological manifestations. Although often diagnosed by primary genetic testing, functional investigation still plays a role in assessing the clinical relevance. In times where whole-exome sequencing (WES) analyses are performed on a regular basis, variants of unknown significance are increasingly discovered in pathways associated with DNA stability and repair, DDR, or cell cycle and proliferation. Since DNA repair defects are associated with increased toxicity towards DNA damaging agents, including radio- or chemotherapy used for anti-cancer treatment or allogeneic hematopoietic stem cell transplantation (HSCT), a rapid diagnosis and correct classification of susceptibility is of utmost importance for outcome and survival of affected patients. Patients affected by (S)CID can be identified by newborn screening using T cell receptor excision circles (TRECs). However, also 50% of AT patients are identified with low TRECs at birth [62].
Although several diagnostic tools are available, not all of them are suitable readouts for all DNA repair deficiencies, or applicable for routine diagnostic tests. In this study, we investigated the use of flow cytometry-based DDR to identify patients affected by radiosensitive immunodeficiencies. DDR biomarkers, such as γH2AX, p-ATM and p-CHK2, were highly useful to identify patients with AT, since ATM plays a major role as a PIKK to activate these factors. Among these biomarkers, p-CHK2 showed the highest selectivity between healthy controls and affected individuals.
In contrast to AT, in which induction of DDR is impaired, NHEJ defects resulted in prolonged repair of DNA DSBs. This was characterized by elevated γH2AX levels at baseline and in response to DNA damage but did not impact on fold induction. In congruence with defects in DNA-PKcs and XLF, also LIG4-deficiency severely impairs repair of DNA DSB, which results in prolonged DDR [63, 64].
Since ARTEMIS is involved in the repair of small fractions of DNA breaks requiring end-processing [55, 65], delayed downregulation of γH2AX becomes apparent only at later time points. However, ARTEMIS deficiency could not be reliably identified by DDR analysis in PBMCs. These findings are controversial to previously reported DNA repair kinetics observed in fibroblasts [58] and Abelson virus-transformed B cell lines [56]. These discrepancies could result from differential DDR and DNA repair capacities of different cell types, which needs to be addressed in future studies. In this study, we observed increased γH2AX responses in NK compared to T lymphocytes. Whereas differential survival responses to DNA damage are well established in lymphocytes [66], DDR has not been systematically compared so far. Furthermore, immunofluorescent staining and scoring of γH2AX foci could be more sensitive than assessment by flow cytometry.
Despite recruitment from multiple centers, small cohorts of RS(S)CID patients could be investigated in this study. To verify results, larger study groups might be needed. Since DDR capacity differs among lymphocyte subsets, we recommend performing analysis on defined subsets such as T and NK lymphocytes.
In assays to quantify recombination activity, including TCRaV7.2 expression [61], NHEJ defects can be identified and separated from other diseases that might affect DDR. A combination of DDR and V(D)J recombination analysis, such as expression of TCRaV7.2, would therefore be a sufficient functional readout for defects of the NHEJ pathway, including ARTEMIS deficiency. Of note, analyses of TCRaV7.2 expression were performed on CD3+ T cells only, and CD161 was not included to discriminate from mucosal-associated invariant T (MAIT) cells [67].
In contrast, flow cytometry-based DDR is not suitable to confirm FA, DC, defects in BER, NER, MMR pathways, and the SWI/SNF complex, or other cancer susceptibility syndromes (Table 3). Many alternative approaches are available, also listed in Table 1. Both colony formation [68] and cellular viability assays have been gold standards to assess cellular DNA repair capacity for the last decades. This approach is highly specific for the identification of DNA repair deficiencies sensitive to IR, such as AT [69]. Alternatively, survival assays can be performed in response to DNA damaging drugs.
Table 3
Application of flow-cytometry based DDR analysis to diagnose DNA repair deficiencies
DNA repair deficiencies
ARTEMIS
LIG4, XLF, DNA-PKcs
AT
FA
ICF
DC
NER, BER, MMR
SWI/SNF
Suitable for flow cytometry-based DDR assay
( +) in combination with V(D)J recombination assays or TCRaV7.2*
 + 
 + 
-
( +) in combination with additional analyses of chromosome stability
-
-
-
*in case MFT has been ruled out
The applicability of flow cytometry-based DDR analysis as a diagnostic tool for various diseases discussed in this study is shown. Alternative approaches are shown in Table 1
FA, Fanconi anemia; ICF, immunodeficiency, centromere instability, facial anomalies; DC, Dyskeratosis congenita; NER, nucleotide excision repair; BER, base excision repair; MMR, mismatch repair; SWI/SNF, SWItch/sucrose non-fermentable
The comet assay is widely used for the analysis of DNA fragmentation by electrophoresis after DNA damage [70], and is an appropriate tool to study DNA repair deficiencies of multiple pathways, including NER, BER [70], AT [69], and FA [71].
Defects in DNA repair may lead to chromosomal instability represented by numerical or structural changes including amplifications, deletions, inversions, and translocations of chromosomal regions. Chromosomes can be studied in multiple ways, which is reviewed in detail by Lepage et al. [72]. Many DNA repair defects are associated with chromosomal instability [73] most commonly observed in FA, AT, Bloom syndrome, and Nijmegen breakage syndrome (NBS), but also in ATLD, ICF and NER syndromes [74], DNA ligase I (LIG1) deficiency [75], and DNA recombinase repair defects [76, 77]. Since chromosomal aberrations are rarely specific for certain diseases, chromosomal studies are rather used for monitoring purposes than for initial diagnostic. Nevertheless, structural abnormalities are highly suggestive for a DNA repair deficiency.
Mononuclear repeat microsatellite sequences are particularly sensitive to transcription errors, making them ideal targets for measuring MSI. Cells with MMR-deficiency accumulate errors, which can create new microsatellite fragments. Microsatellites can be sequenced, analyzed by fluorescent PCR, or immunofluorescent staining [78] to provide evidence for MSI.
Short telomeres are a diagnostic criterion for DC [42], but can also be found in other DNA repair deficiencies including LIG4 deficiency and Dubowitz syndrome [79].
The DDR impacts on the G1/S, intra-S, and G2/M cell cycle checkpoints that allow time for DNA repair before replication and cell division [80]. Cell cycle response to DNA damage can be a good readout for many DNA repair defects that involve proteins operating in cell cycle regulation. Failure to inhibit DNA synthesis after DNA damage (G1/S and intra-S checkpoint) is a hallmark of ATM-deficient cells [81]. A prolonged cell cycle arrest at the G2/M checkpoint can be observed in NHEJ-deficient cells [82]. Treatment with alkylating nitrogen mustard (NM) or mitomycin C (MMC) results in cell cycle arrest at G2/M in cells with defects in the FA pathway, which is frequently used to diagnose FA [83].
Furthermore, additional DDR biomarkers altered by phosphorylation can be used to study DDR and DNA repair capacity. Impaired phosphorylation of structural maintenance of chromosome 1 (SMC1) has been reported in patients with AT and was also reduced in heterozygous carriers of ATM mutations [84]. Rosen et al. reported the use of p-DNA-PKcs, p-53BP1, p-RPA2/32, p-BRCA1, p-p53, and p21 as additional DDR biomarkers in ATM-, BRCA1-, and BRCA2-deficient cell lines treated with etoposide [85].
Compared to available diagnostic tests, flow cytometry-based assays can be performed on peripheral blood cells and provide quick results. In contrast to cell cycle and cell survival assays, DDR is not dependent on proliferative response to mitogen stimulation, which can be a serious limitation in cellular material obtained from patients with primary immunodeficiencies. However, more biomarkers need to be identified to cover the heterogeneity of DNA repair deficiencies. Of note, DDR is not affected in HR- or FA-related defects, although DNA repair kinetics may be altered.
Genetic defects affecting DNA stability and repair can lead to overlapping presentations of immunodeficiency, bone marrow failure, and cancer susceptibility, and functional diagnostics can be challenging. Identification of diagnostic algorithms with the use of specific analysis tools can help to discriminate affected pathways; however, up to now, one test alone will not be able to cover the complete spectrum.

Acknowledgements

The authors would like to thank Dilek Dayanakli, Eva-Maria Rump, and Sarah Radecke for their technical and advisory support.

Declarations

Ethics Approval

This study was approved by the ethic committees of Ulm University (407/16), Technical University of Dresden (TUD) (BO-EK-213052020), and Hannover Medical School (MHH) (9492-BO-K-2020), Germany.
Informed consent was obtained from all individual participants, or their legal guardians, included in the study.
All participants, or their legal guardians, signed informed consent regarding publishing their anonymized data.

Conflict of Interest

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Wang XS, Lee BJ, Zha S. The recent advances in non-homologous end-joining through the lens of lymphocyte development. DNA Repair (Amst). 2020;94:102874.PubMedPubMedCentralCrossRef Wang XS, Lee BJ, Zha S. The recent advances in non-homologous end-joining through the lens of lymphocyte development. DNA Repair (Amst). 2020;94:102874.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Wu Q. Structural mechanism of DNA-end synapsis in the non-homologous end joining pathway for repairing double-strand breaks: bridge over troubled ends. Biochem Soc Trans. 2019;47(6):1609–19.PubMedCrossRef Wu Q. Structural mechanism of DNA-end synapsis in the non-homologous end joining pathway for repairing double-strand breaks: bridge over troubled ends. Biochem Soc Trans. 2019;47(6):1609–19.PubMedCrossRef
5.
Zurück zum Zitat Slatter MA, Gennery AR. Update on DNA-double strand break repair defects in combined primary immunodeficiency. Curr Allergy Asthma Rep. 2020;20(10):57.PubMedPubMedCentralCrossRef Slatter MA, Gennery AR. Update on DNA-double strand break repair defects in combined primary immunodeficiency. Curr Allergy Asthma Rep. 2020;20(10):57.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Moshous D, Callebaut I, de Chasseval R, Corneo B, Cavazzana-Calvo M, Le Deist F, et al. Artemis, a novel DNA double-strand break repair/V(D)J recombination protein, is mutated in human severe combined immune deficiency. Cell. 2001;105(2):177–86.PubMedCrossRef Moshous D, Callebaut I, de Chasseval R, Corneo B, Cavazzana-Calvo M, Le Deist F, et al. Artemis, a novel DNA double-strand break repair/V(D)J recombination protein, is mutated in human severe combined immune deficiency. Cell. 2001;105(2):177–86.PubMedCrossRef
7.
Zurück zum Zitat Volk T, Pannicke U, Reisli I, Bulashevska A, Ritter J, Bjorkman A, et al. DCLRE1C (ARTEMIS) mutations causing phenotypes ranging from atypical severe combined immunodeficiency to mere antibody deficiency. Hum Mol Genet. 2015;24(25):7361–72.PubMedPubMedCentralCrossRef Volk T, Pannicke U, Reisli I, Bulashevska A, Ritter J, Bjorkman A, et al. DCLRE1C (ARTEMIS) mutations causing phenotypes ranging from atypical severe combined immunodeficiency to mere antibody deficiency. Hum Mol Genet. 2015;24(25):7361–72.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat O’Driscoll M, Cerosaletti KM, Girard PM, Dai Y, Stumm M, Kysela B, et al. DNA ligase IV mutations identified in patients exhibiting developmental delay and immunodeficiency. Mol Cell. 2001;8(6):1175–85.PubMedCrossRef O’Driscoll M, Cerosaletti KM, Girard PM, Dai Y, Stumm M, Kysela B, et al. DNA ligase IV mutations identified in patients exhibiting developmental delay and immunodeficiency. Mol Cell. 2001;8(6):1175–85.PubMedCrossRef
9.
Zurück zum Zitat Murray JE, Bicknell LS, Yigit G, Duker AL, van Kogelenberg M, Haghayegh S, et al. Extreme growth failure is a common presentation of ligase IV deficiency. Hum Mutat. 2014;35(1):76–85.PubMedCrossRef Murray JE, Bicknell LS, Yigit G, Duker AL, van Kogelenberg M, Haghayegh S, et al. Extreme growth failure is a common presentation of ligase IV deficiency. Hum Mutat. 2014;35(1):76–85.PubMedCrossRef
10.
Zurück zum Zitat Buck D, Moshous D, de Chasseval R, Ma Y, le Deist F, Cavazzana-Calvo M, et al. Severe combined immunodeficiency and microcephaly in siblings with hypomorphic mutations in DNA ligase IV. Eur J Immunol. 2006;36(1):224–35.PubMedCrossRef Buck D, Moshous D, de Chasseval R, Ma Y, le Deist F, Cavazzana-Calvo M, et al. Severe combined immunodeficiency and microcephaly in siblings with hypomorphic mutations in DNA ligase IV. Eur J Immunol. 2006;36(1):224–35.PubMedCrossRef
11.
Zurück zum Zitat Buck D, Malivert L, de Chasseval R, Barraud A, Fondaneche MC, Sanal O, et al. Cernunnos, a novel nonhomologous end-joining factor, is mutated in human immunodeficiency with microcephaly. Cell. 2006;124(2):287–99.PubMedCrossRef Buck D, Malivert L, de Chasseval R, Barraud A, Fondaneche MC, Sanal O, et al. Cernunnos, a novel nonhomologous end-joining factor, is mutated in human immunodeficiency with microcephaly. Cell. 2006;124(2):287–99.PubMedCrossRef
12.
Zurück zum Zitat Dutrannoy V, Demuth I, Baumann U, Schindler D, Konrat K, Neitzel H, et al. Clinical variability and novel mutations in the NHEJ1 gene in patients with a Nijmegen breakage syndrome-like phenotype. Hum Mutat. 2010;31(9):1059–68.PubMedCrossRef Dutrannoy V, Demuth I, Baumann U, Schindler D, Konrat K, Neitzel H, et al. Clinical variability and novel mutations in the NHEJ1 gene in patients with a Nijmegen breakage syndrome-like phenotype. Hum Mutat. 2010;31(9):1059–68.PubMedCrossRef
13.
Zurück zum Zitat van der Burg M, van Dongen JJ, van Gent DC. DNA-PKcs deficiency in human: long predicted, finally found. Curr Opin Allergy Clin Immunol. 2009;9(6):503–9.PubMedCrossRef van der Burg M, van Dongen JJ, van Gent DC. DNA-PKcs deficiency in human: long predicted, finally found. Curr Opin Allergy Clin Immunol. 2009;9(6):503–9.PubMedCrossRef
14.
Zurück zum Zitat Woodbine L, Neal JA, Sasi NK, Shimada M, Deem K, Coleman H, et al. PRKDC mutations in a SCID patient with profound neurological abnormalities. J Clin Invest. 2013;123(7):2969–80.PubMedPubMedCentralCrossRef Woodbine L, Neal JA, Sasi NK, Shimada M, Deem K, Coleman H, et al. PRKDC mutations in a SCID patient with profound neurological abnormalities. J Clin Invest. 2013;123(7):2969–80.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Mathieu AL, Verronese E, Rice GI, Fouyssac F, Bertrand Y, Picard C, et al. PRKDC mutations associated with immunodeficiency, granuloma, and autoimmune regulator-dependent autoimmunity. J Allergy Clin Immunol. 2015;135(6):1578-88.e5.PubMedPubMedCentralCrossRef Mathieu AL, Verronese E, Rice GI, Fouyssac F, Bertrand Y, Picard C, et al. PRKDC mutations associated with immunodeficiency, granuloma, and autoimmune regulator-dependent autoimmunity. J Allergy Clin Immunol. 2015;135(6):1578-88.e5.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Esenboga S, Akal C, Karaatmaca B, Erman B, Dogan S, Orhan D, et al. Two siblings with PRKDC defect who presented with cutaneous granulomas and review of the literature. Clin Immunol. 2018;197:1–5.PubMedCrossRef Esenboga S, Akal C, Karaatmaca B, Erman B, Dogan S, Orhan D, et al. Two siblings with PRKDC defect who presented with cutaneous granulomas and review of the literature. Clin Immunol. 2018;197:1–5.PubMedCrossRef
17.
Zurück zum Zitat Bee L, Nasca A, Zanolini A, Cendron F, d’Adamo P, Costa R, et al. A nonsense mutation of human XRCC4 is associated with adult-onset progressive encephalocardiomyopathy. EMBO Mol Med. 2015;7(7):918–29.PubMedPubMedCentralCrossRef Bee L, Nasca A, Zanolini A, Cendron F, d’Adamo P, Costa R, et al. A nonsense mutation of human XRCC4 is associated with adult-onset progressive encephalocardiomyopathy. EMBO Mol Med. 2015;7(7):918–29.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Guo C, Nakazawa Y, Woodbine L, Bjorkman A, Shimada M, Fawcett H, et al. XRCC4 deficiency in human subjects causes a marked neurological phenotype but no overt immunodeficiency. J Allergy Clin Immunol. 2015;136(4):1007–17.PubMedCrossRef Guo C, Nakazawa Y, Woodbine L, Bjorkman A, Shimada M, Fawcett H, et al. XRCC4 deficiency in human subjects causes a marked neurological phenotype but no overt immunodeficiency. J Allergy Clin Immunol. 2015;136(4):1007–17.PubMedCrossRef
19.
Zurück zum Zitat Rosin N, Elcioglu NH, Beleggia F, Isguven P, Altmuller J, Thiele H, et al. Mutations in XRCC4 cause primary microcephaly, short stature and increased genomic instability. Hum Mol Genet. 2015;24(13):3708–17.PubMed Rosin N, Elcioglu NH, Beleggia F, Isguven P, Altmuller J, Thiele H, et al. Mutations in XRCC4 cause primary microcephaly, short stature and increased genomic instability. Hum Mol Genet. 2015;24(13):3708–17.PubMed
20.
Zurück zum Zitat Murray JE, van der Burg M, H IJ, Carroll P, Wu Q, Ochi T, et al. Mutations in the NHEJ component XRCC4 cause primordial dwarfism. Am J Hum Genet. 2015;96(3):412–24. Murray JE, van der Burg M, H IJ, Carroll P, Wu Q, Ochi T, et al. Mutations in the NHEJ component XRCC4 cause primordial dwarfism. Am J Hum Genet. 2015;96(3):412–24.
21.
Zurück zum Zitat Xing M, Yang M, Huo W, Feng F, Wei L, Jiang W, et al. Interactome analysis identifies a new paralogue of XRCC4 in non-homologous end joining DNA repair pathway. Nat Commun. 2015;6:6233.PubMedCrossRef Xing M, Yang M, Huo W, Feng F, Wei L, Jiang W, et al. Interactome analysis identifies a new paralogue of XRCC4 in non-homologous end joining DNA repair pathway. Nat Commun. 2015;6:6233.PubMedCrossRef
22.
Zurück zum Zitat Ochi T, Blackford AN, Coates J, Jhujh S, Mehmood S, Tamura N, et al. DNA repair. PAXX, a paralog of XRCC4 and XLF, interacts with Ku to promote DNA double-strand break repair. Science. 2015;347(6218):185–8. Ochi T, Blackford AN, Coates J, Jhujh S, Mehmood S, Tamura N, et al. DNA repair. PAXX, a paralog of XRCC4 and XLF, interacts with Ku to promote DNA double-strand break repair. Science. 2015;347(6218):185–8.
23.
Zurück zum Zitat Kumar V, Alt FW, Frock RL. PAXX and XLF DNA repair factors are functionally redundant in joining DNA breaks in a G1-arrested progenitor B-cell line. Proc Natl Acad Sci U S A. 2016;113(38):10619–24.PubMedPubMedCentralCrossRef Kumar V, Alt FW, Frock RL. PAXX and XLF DNA repair factors are functionally redundant in joining DNA breaks in a G1-arrested progenitor B-cell line. Proc Natl Acad Sci U S A. 2016;113(38):10619–24.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat van den Bosch M, Bree RT, Lowndes NF. The MRN complex: coordinating and mediating the response to broken chromosomes. EMBO Rep. 2003;4(9):844–9.PubMedPubMedCentralCrossRef van den Bosch M, Bree RT, Lowndes NF. The MRN complex: coordinating and mediating the response to broken chromosomes. EMBO Rep. 2003;4(9):844–9.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat McGrath-Morrow SA, Rothblum-Oviatt CC, Wright J, Schlechter H, Lefton-Greif MA, Natale VA, et al. Multidisciplinary management of ataxia telangiectasia: current perspectives. J Multidiscip Healthc. 2021;14:1637–44.PubMedPubMedCentralCrossRef McGrath-Morrow SA, Rothblum-Oviatt CC, Wright J, Schlechter H, Lefton-Greif MA, Natale VA, et al. Multidisciplinary management of ataxia telangiectasia: current perspectives. J Multidiscip Healthc. 2021;14:1637–44.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Weitering TJ, Takada S, Weemaes CMR, van Schouwenburg PA, van der Burg M. ATM: translating the DNA damage response to adaptive immunity. Trends Immunol. 2021;42(4):350–65.PubMedCrossRef Weitering TJ, Takada S, Weemaes CMR, van Schouwenburg PA, van der Burg M. ATM: translating the DNA damage response to adaptive immunity. Trends Immunol. 2021;42(4):350–65.PubMedCrossRef
28.
Zurück zum Zitat Stewart GS, Maser RS, Stankovic T, Bressan DA, Kaplan MI, Jaspers NG, et al. The DNA double-strand break repair gene hMRE11 is mutated in individuals with an ataxia-telangiectasia-like disorder. Cell. 1999;99(6):577–87.PubMedCrossRef Stewart GS, Maser RS, Stankovic T, Bressan DA, Kaplan MI, Jaspers NG, et al. The DNA double-strand break repair gene hMRE11 is mutated in individuals with an ataxia-telangiectasia-like disorder. Cell. 1999;99(6):577–87.PubMedCrossRef
29.
Zurück zum Zitat Fievet A, Bellanger D, Valence S, Mobuchon L, Afenjar A, Giuliano F, et al. Three new cases of ataxia-telangiectasia-like disorder: no impairment of the ATM pathway, but S-phase checkpoint defect. Hum Mutat. 2019;40(10):1690–9.PubMedCrossRef Fievet A, Bellanger D, Valence S, Mobuchon L, Afenjar A, Giuliano F, et al. Three new cases of ataxia-telangiectasia-like disorder: no impairment of the ATM pathway, but S-phase checkpoint defect. Hum Mutat. 2019;40(10):1690–9.PubMedCrossRef
30.
Zurück zum Zitat Varon R, Vissinga C, Platzer M, Cerosaletti KM, Chrzanowska KH, Saar K, et al. Nibrin, a novel DNA double-strand break repair protein, is mutated in Nijmegen breakage syndrome. Cell. 1998;93(3):467–76.PubMedCrossRef Varon R, Vissinga C, Platzer M, Cerosaletti KM, Chrzanowska KH, Saar K, et al. Nibrin, a novel DNA double-strand break repair protein, is mutated in Nijmegen breakage syndrome. Cell. 1998;93(3):467–76.PubMedCrossRef
31.
Zurück zum Zitat Wolska-Kusnierz B, Gregorek H, Chrzanowska K, Piatosa B, Pietrucha B, Heropolitanska-Pliszka E, et al. Nijmegen breakage syndrome: clinical and immunological features, long-term outcome and treatment options — a retrospective analysis. J Clin Immunol. 2015;35(6):538–49.PubMedCrossRef Wolska-Kusnierz B, Gregorek H, Chrzanowska K, Piatosa B, Pietrucha B, Heropolitanska-Pliszka E, et al. Nijmegen breakage syndrome: clinical and immunological features, long-term outcome and treatment options — a retrospective analysis. J Clin Immunol. 2015;35(6):538–49.PubMedCrossRef
32.
Zurück zum Zitat Waltes R, Kalb R, Gatei M, Kijas AW, Stumm M, Sobeck A, et al. Human RAD50 deficiency in a Nijmegen breakage syndrome-like disorder. Am J Hum Genet. 2009;84(5):605–16.PubMedPubMedCentralCrossRef Waltes R, Kalb R, Gatei M, Kijas AW, Stumm M, Sobeck A, et al. Human RAD50 deficiency in a Nijmegen breakage syndrome-like disorder. Am J Hum Genet. 2009;84(5):605–16.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Ragamin A, Yigit G, Bousset K, Beleggia F, Verheijen FW, de Wit MY, et al. Human RAD50 deficiency: confirmation of a distinctive phenotype. Am J Med Genet A. 2020;182(6):1378–86.PubMedPubMedCentralCrossRef Ragamin A, Yigit G, Bousset K, Beleggia F, Verheijen FW, de Wit MY, et al. Human RAD50 deficiency: confirmation of a distinctive phenotype. Am J Med Genet A. 2020;182(6):1378–86.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Cunniff C, Bassetti JA, Ellis NA. Bloom’s syndrome: clinical spectrum, molecular pathogenesis, and cancer predisposition. Mol Syndromol. 2017;8(1):4–23.PubMedCrossRef Cunniff C, Bassetti JA, Ellis NA. Bloom’s syndrome: clinical spectrum, molecular pathogenesis, and cancer predisposition. Mol Syndromol. 2017;8(1):4–23.PubMedCrossRef
35.
Zurück zum Zitat Schoenaker MHD, Henriet SS, Zonderland J, van Deuren M, Pan-Hammarstrom Q, Posthumus-van Sluijs SJ, et al. Immunodeficiency in Bloom’s Syndrome. J Clin Immunol. 2018;38(1):35–44.PubMedCrossRef Schoenaker MHD, Henriet SS, Zonderland J, van Deuren M, Pan-Hammarstrom Q, Posthumus-van Sluijs SJ, et al. Immunodeficiency in Bloom’s Syndrome. J Clin Immunol. 2018;38(1):35–44.PubMedCrossRef
36.
Zurück zum Zitat Garcia-de-Teresa B, Rodriguez A, Frias S. Chromosome instability in Fanconi anemia: from breaks to phenotypic consequences. Genes (Basel). 2020;11(12). Garcia-de-Teresa B, Rodriguez A, Frias S. Chromosome instability in Fanconi anemia: from breaks to phenotypic consequences. Genes (Basel). 2020;11(12).
37.
Zurück zum Zitat Myers KC, Sauter S, Zhang X, Bleesing JJ, Davies SM, Wells SI, et al. Impaired immune function in children and adults with Fanconi anemia. Pediatr Blood Cancer. 2017;64(11). Myers KC, Sauter S, Zhang X, Bleesing JJ, Davies SM, Wells SI, et al. Impaired immune function in children and adults with Fanconi anemia. Pediatr Blood Cancer. 2017;64(11).
38.
Zurück zum Zitat Krasikova Y, Rechkunova N, Lavrik O. Nucleotide excision repair: from molecular defects to neurological abnormalities. Int J Mol Sci. 2021;22(12). Krasikova Y, Rechkunova N, Lavrik O. Nucleotide excision repair: from molecular defects to neurological abnormalities. Int J Mol Sci. 2021;22(12).
39.
Zurück zum Zitat Stratigopoulou M, van Dam TP, Guikema JEJ. Base excision repair in the immune system: small DNA lesions with big consequences. Front Immunol. 2020;11:1084.PubMedPubMedCentralCrossRef Stratigopoulou M, van Dam TP, Guikema JEJ. Base excision repair in the immune system: small DNA lesions with big consequences. Front Immunol. 2020;11:1084.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat H IJ, van Schouwenburg PA, Pico-Knijnenburg I, Loeffen J, Brugieres L, Driessen GJ, et al. Repertoire sequencing of B cells elucidates the role of UNG and mismatch repair proteins in somatic hypermutation in humans. Front Immunol. 2019;10:1913. H IJ, van Schouwenburg PA, Pico-Knijnenburg I, Loeffen J, Brugieres L, Driessen GJ, et al. Repertoire sequencing of B cells elucidates the role of UNG and mismatch repair proteins in somatic hypermutation in humans. Front Immunol. 2019;10:1913.
42.
Zurück zum Zitat Fernandez Garcia MS, Teruya-Feldstein J. The diagnosis and treatment of dyskeratosis congenita: a review. J Blood Med. 2014;5:157–67.PubMedPubMedCentral Fernandez Garcia MS, Teruya-Feldstein J. The diagnosis and treatment of dyskeratosis congenita: a review. J Blood Med. 2014;5:157–67.PubMedPubMedCentral
43.
Zurück zum Zitat Kiaee F, Zaki-Dizaji M, Hafezi N, Almasi-Hashiani A, Hamedifar H, Sabzevari A, et al. Clinical, immunologic, and molecular spectrum of patients with immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome: a systematic review. Endocr Metab Immune Disord Drug Targets. 2020. Kiaee F, Zaki-Dizaji M, Hafezi N, Almasi-Hashiani A, Hamedifar H, Sabzevari A, et al. Clinical, immunologic, and molecular spectrum of patients with immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome: a systematic review. Endocr Metab Immune Disord Drug Targets. 2020.
44.
Zurück zum Zitat Jyonouchi S, Forbes L, Ruchelli E, Sullivan KE. Dyskeratosis congenita: a combined immunodeficiency with broad clinical spectrum—a single-center pediatric experience. Pediatr Allergy Immunol. 2011;22(3):313–9.PubMedCrossRef Jyonouchi S, Forbes L, Ruchelli E, Sullivan KE. Dyskeratosis congenita: a combined immunodeficiency with broad clinical spectrum—a single-center pediatric experience. Pediatr Allergy Immunol. 2011;22(3):313–9.PubMedCrossRef
45.
Zurück zum Zitat Allenspach EJ, Bellodi C, Jeong D, Kopmar N, Nakamura T, Ochs HD, et al. Common variable immunodeficiency as the initial presentation of dyskeratosis congenita. J Allergy Clin Immunol. 2013;132(1):223–6.PubMedPubMedCentralCrossRef Allenspach EJ, Bellodi C, Jeong D, Kopmar N, Nakamura T, Ochs HD, et al. Common variable immunodeficiency as the initial presentation of dyskeratosis congenita. J Allergy Clin Immunol. 2013;132(1):223–6.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Helfricht A, Thijssen PE, Rother MB, Shah RG, Du L, Takada S, et al. Loss of ZBTB24 impairs nonhomologous end-joining and class-switch recombination in patients with ICF syndrome. J Exp Med. 2020;217(11). Helfricht A, Thijssen PE, Rother MB, Shah RG, Du L, Takada S, et al. Loss of ZBTB24 impairs nonhomologous end-joining and class-switch recombination in patients with ICF syndrome. J Exp Med. 2020;217(11).
47.
Zurück zum Zitat He Y, Ren J, Xu X, Ni K, Schwader A, Finney R, et al. Lsh/HELLS is required for B lymphocyte development and immunoglobulin class switch recombination. Proc Natl Acad Sci U S A. 2020;117(33):20100–8.PubMedPubMedCentralCrossRef He Y, Ren J, Xu X, Ni K, Schwader A, Finney R, et al. Lsh/HELLS is required for B lymphocyte development and immunoglobulin class switch recombination. Proc Natl Acad Sci U S A. 2020;117(33):20100–8.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Unoki M, Funabiki H, Velasco G, Francastel C, Sasaki H. CDCA7 and HELLS mutations undermine nonhomologous end joining in centromeric instability syndrome. J Clin Invest. 2019;129(1):78–92.PubMedCrossRef Unoki M, Funabiki H, Velasco G, Francastel C, Sasaki H. CDCA7 and HELLS mutations undermine nonhomologous end joining in centromeric instability syndrome. J Clin Invest. 2019;129(1):78–92.PubMedCrossRef
49.
Zurück zum Zitat Moon SH, Nguyen TA, Darlington Y, Lu X, Donehower LA. Dephosphorylation of gamma-H2AX by WIP1: an important homeostatic regulatory event in DNA repair and cell cycle control. Cell Cycle. 2010;9(11):2092–6.PubMedCrossRef Moon SH, Nguyen TA, Darlington Y, Lu X, Donehower LA. Dephosphorylation of gamma-H2AX by WIP1: an important homeostatic regulatory event in DNA repair and cell cycle control. Cell Cycle. 2010;9(11):2092–6.PubMedCrossRef
50.
Zurück zum Zitat Chowdhury D, Keogh MC, Ishii H, Peterson CL, Buratowski S, Lieberman J. gamma-H2AX dephosphorylation by protein phosphatase 2A facilitates DNA double-strand break repair. Mol Cell. 2005;20(5):801–9.PubMedCrossRef Chowdhury D, Keogh MC, Ishii H, Peterson CL, Buratowski S, Lieberman J. gamma-H2AX dephosphorylation by protein phosphatase 2A facilitates DNA double-strand break repair. Mol Cell. 2005;20(5):801–9.PubMedCrossRef
51.
Zurück zum Zitat Chowdhury D, Xu X, Zhong X, Ahmed F, Zhong J, Liao J, et al. A PP4-phosphatase complex dephosphorylates gamma-H2AX generated during DNA replication. Mol Cell. 2008;31(1):33–46.PubMedPubMedCentralCrossRef Chowdhury D, Xu X, Zhong X, Ahmed F, Zhong J, Liao J, et al. A PP4-phosphatase complex dephosphorylates gamma-H2AX generated during DNA replication. Mol Cell. 2008;31(1):33–46.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273(10):5858–68.PubMedCrossRef Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem. 1998;273(10):5858–68.PubMedCrossRef
53.
Zurück zum Zitat Muslimovic A, Ismail IH, Gao Y, Hammarsten O. An optimized method for measurement of gamma-H2AX in blood mononuclear and cultured cells. Nat Protoc. 2008;3(7):1187–93.PubMedCrossRef Muslimovic A, Ismail IH, Gao Y, Hammarsten O. An optimized method for measurement of gamma-H2AX in blood mononuclear and cultured cells. Nat Protoc. 2008;3(7):1187–93.PubMedCrossRef
54.
Zurück zum Zitat Lobrich M, Shibata A, Beucher A, Fisher A, Ensminger M, Goodarzi AA, et al. gammaH2AX foci analysis for monitoring DNA double-strand break repair: strengths, limitations and optimization. Cell Cycle. 2010;9(4):662–9.PubMedCrossRef Lobrich M, Shibata A, Beucher A, Fisher A, Ensminger M, Goodarzi AA, et al. gammaH2AX foci analysis for monitoring DNA double-strand break repair: strengths, limitations and optimization. Cell Cycle. 2010;9(4):662–9.PubMedCrossRef
55.
Zurück zum Zitat Lobrich M, Jeggo P. A process of resection-dependent nonhomologous end joining involving the Goddess Artemis. Trends Biochem Sci. 2017;42(9):690–701.PubMedPubMedCentralCrossRef Lobrich M, Jeggo P. A process of resection-dependent nonhomologous end joining involving the Goddess Artemis. Trends Biochem Sci. 2017;42(9):690–701.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Felgentreff K, Lee YN, Frugoni F, Du L, van der Burg M, Giliani S, et al. Functional analysis of naturally occurring DCLRE1C mutations and correlation with the clinical phenotype of ARTEMIS deficiency. J Allergy Clin Immunol. 2015;136(1):140–50 e7. Felgentreff K, Lee YN, Frugoni F, Du L, van der Burg M, Giliani S, et al. Functional analysis of naturally occurring DCLRE1C mutations and correlation with the clinical phenotype of ARTEMIS deficiency. J Allergy Clin Immunol. 2015;136(1):140–50 e7.
57.
Zurück zum Zitat Lobachevsky P, Woodbine L, Hsiao KC, Choo S, Fraser C, Gray P, et al. Evaluation of severe combined immunodeficiency and combined immunodeficiency pediatric patients on the basis of cellular radiosensitivity. J Mol Diagn. 2015;17(5):560–75.PubMedPubMedCentralCrossRef Lobachevsky P, Woodbine L, Hsiao KC, Choo S, Fraser C, Gray P, et al. Evaluation of severe combined immunodeficiency and combined immunodeficiency pediatric patients on the basis of cellular radiosensitivity. J Mol Diagn. 2015;17(5):560–75.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Martin OA, Ivashkevich A, Choo S, Woodbine L, Jeggo PA, Martin RF, et al. Statistical analysis of kinetics, distribution and co-localisation of DNA repair foci in irradiated cells: cell cycle effect and implications for prediction of radiosensitivity. DNA Repair (Amst). 2013;12(10):844–55.CrossRef Martin OA, Ivashkevich A, Choo S, Woodbine L, Jeggo PA, Martin RF, et al. Statistical analysis of kinetics, distribution and co-localisation of DNA repair foci in irradiated cells: cell cycle effect and implications for prediction of radiosensitivity. DNA Repair (Amst). 2013;12(10):844–55.CrossRef
59.
Zurück zum Zitat Felgentreff K, Du L, Weinacht KG, Dobbs K, Bartish M, Giliani S, et al. Differential role of nonhomologous end joining factors in the generation, DNA damage response, and myeloid differentiation of human induced pluripotent stem cells. Proc Natl Acad Sci U S A. 2014;111(24):8889–94.PubMedPubMedCentralCrossRef Felgentreff K, Du L, Weinacht KG, Dobbs K, Bartish M, Giliani S, et al. Differential role of nonhomologous end joining factors in the generation, DNA damage response, and myeloid differentiation of human induced pluripotent stem cells. Proc Natl Acad Sci U S A. 2014;111(24):8889–94.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Leskovac A, Vujic D, Guc-Scekic M, Petrovic S, Joksic I, Slijepcevic P, et al. Fanconi anemia is characterized by delayed repair kinetics of DNA double-strand breaks. Tohoku J Exp Med. 2010;221(1):69–76.PubMedCrossRef Leskovac A, Vujic D, Guc-Scekic M, Petrovic S, Joksic I, Slijepcevic P, et al. Fanconi anemia is characterized by delayed repair kinetics of DNA double-strand breaks. Tohoku J Exp Med. 2010;221(1):69–76.PubMedCrossRef
61.
Zurück zum Zitat Berland A, Rosain J, Kaltenbach S, Allain V, Mahlaoui N, Melki I, et al. PROMIDISalpha: A T-cell receptor alpha signature associated with immunodeficiencies caused by V(D)J recombination defects. J Allergy Clin Immunol. 2019;143(1):325–34 e2. Berland A, Rosain J, Kaltenbach S, Allain V, Mahlaoui N, Melki I, et al. PROMIDISalpha: A T-cell receptor alpha signature associated with immunodeficiencies caused by V(D)J recombination defects. J Allergy Clin Immunol. 2019;143(1):325–34 e2.
62.
Zurück zum Zitat Mallott J, Kwan A, Church J, Gonzalez-Espinosa D, Lorey F, Tang LF, et al. Newborn screening for SCID identifies patients with ataxia telangiectasia. J Clin Immunol. 2013;33(3):540–9.PubMedCrossRef Mallott J, Kwan A, Church J, Gonzalez-Espinosa D, Lorey F, Tang LF, et al. Newborn screening for SCID identifies patients with ataxia telangiectasia. J Clin Immunol. 2013;33(3):540–9.PubMedCrossRef
63.
Zurück zum Zitat Felgentreff K, Baxi SN, Lee YN, Dobbs K, Henderson LA, Csomos K, et al. Ligase-4 deficiency causes distinctive immune abnormalities in asymptomatic individuals. J Clin Immunol. 2016;36(4):341–53.PubMedPubMedCentralCrossRef Felgentreff K, Baxi SN, Lee YN, Dobbs K, Henderson LA, Csomos K, et al. Ligase-4 deficiency causes distinctive immune abnormalities in asymptomatic individuals. J Clin Immunol. 2016;36(4):341–53.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Buchbinder D, Smith MJ, Kawahara M, Cowan MJ, Buzby JS, Abraham RS. Application of a radiosensitivity flow assay in a patient with DNA ligase 4 deficiency. Blood Adv. 2018;2(15):1828–32.PubMedPubMedCentralCrossRef Buchbinder D, Smith MJ, Kawahara M, Cowan MJ, Buzby JS, Abraham RS. Application of a radiosensitivity flow assay in a patient with DNA ligase 4 deficiency. Blood Adv. 2018;2(15):1828–32.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Riballo E, Kuhne M, Rief N, Doherty A, Smith GC, Recio MJ, et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci. Mol Cell. 2004;16(5):715–24.PubMedCrossRef Riballo E, Kuhne M, Rief N, Doherty A, Smith GC, Recio MJ, et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci. Mol Cell. 2004;16(5):715–24.PubMedCrossRef
66.
Zurück zum Zitat Wilkins RC, Wilkinson D, Maharaj HP, Bellier PV, Cybulski MB, McLean JR. Differential apoptotic response to ionizing radiation in subpopulations of human white blood cells. Mutat Res. 2002;513(1–2):27–36.PubMedCrossRef Wilkins RC, Wilkinson D, Maharaj HP, Bellier PV, Cybulski MB, McLean JR. Differential apoptotic response to ionizing radiation in subpopulations of human white blood cells. Mutat Res. 2002;513(1–2):27–36.PubMedCrossRef
67.
Zurück zum Zitat Kurioka A, Jahun AS, Hannaway RF, Walker LJ, Fergusson JR, Sverremark-Ekstrom E, et al. Shared and distinct phenotypes and functions of human CD161++ Valpha7.2+ T cell subsets. Front Immunol. 2017;8:1031. Kurioka A, Jahun AS, Hannaway RF, Walker LJ, Fergusson JR, Sverremark-Ekstrom E, et al. Shared and distinct phenotypes and functions of human CD161++ Valpha7.2+ T cell subsets. Front Immunol. 2017;8:1031.
68.
Zurück zum Zitat Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9.PubMedCrossRef Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9.PubMedCrossRef
69.
Zurück zum Zitat Nahas SA, Davies R, Fike F, Nakamura K, Du L, Kayali R, et al. Comprehensive profiling of radiosensitive human cell lines with DNA damage response assays identifies the neutral comet assay as a potential surrogate for clonogenic survival. Radiat Res. 2012;177(2):176–86.PubMedCrossRef Nahas SA, Davies R, Fike F, Nakamura K, Du L, Kayali R, et al. Comprehensive profiling of radiosensitive human cell lines with DNA damage response assays identifies the neutral comet assay as a potential surrogate for clonogenic survival. Radiat Res. 2012;177(2):176–86.PubMedCrossRef
70.
Zurück zum Zitat Azqueta A, Slyskova J, Langie SA, O’Neill Gaivao I, Collins A. Comet assay to measure DNA repair: approach and applications. Front Genet. 2014;5:288.PubMedPubMedCentralCrossRef Azqueta A, Slyskova J, Langie SA, O’Neill Gaivao I, Collins A. Comet assay to measure DNA repair: approach and applications. Front Genet. 2014;5:288.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Djuzenova CS, Flentje M. Characterization of Fanconi anemia fibroblasts in terms of clonogenic survival and DNA damage assessed by the Comet assay. Med Sci Monit. 2002;8(10):BR421–30. Djuzenova CS, Flentje M. Characterization of Fanconi anemia fibroblasts in terms of clonogenic survival and DNA damage assessed by the Comet assay. Med Sci Monit. 2002;8(10):BR421–30.
72.
Zurück zum Zitat Lepage CC, Morden CR, Palmer MCL, Nachtigal MW, McManus KJ. Detecting chromosome instability in cancer: approaches to resolve cell-to-cell heterogeneity. Cancers (Basel). 2019;11(2). Lepage CC, Morden CR, Palmer MCL, Nachtigal MW, McManus KJ. Detecting chromosome instability in cancer: approaches to resolve cell-to-cell heterogeneity. Cancers (Basel). 2019;11(2).
73.
Zurück zum Zitat Taylor AMR, Rothblum-Oviatt C, Ellis NA, Hickson ID, Meyer S, Crawford TO, et al. Chromosome instability syndromes. Nat Rev Dis Primers. 2019;5(1):64.PubMedCrossRef Taylor AMR, Rothblum-Oviatt C, Ellis NA, Hickson ID, Meyer S, Crawford TO, et al. Chromosome instability syndromes. Nat Rev Dis Primers. 2019;5(1):64.PubMedCrossRef
75.
Zurück zum Zitat Liddiard K, Ruis B, Kan Y, Cleal K, Ashelford KE, Hendrickson EA, et al. DNA Ligase 1 is an essential mediator of sister chromatid telomere fusions in G2 cell cycle phase. Nucleic Acids Res. 2019;47(5):2402–24.PubMedCrossRef Liddiard K, Ruis B, Kan Y, Cleal K, Ashelford KE, Hendrickson EA, et al. DNA Ligase 1 is an essential mediator of sister chromatid telomere fusions in G2 cell cycle phase. Nucleic Acids Res. 2019;47(5):2402–24.PubMedCrossRef
77.
Zurück zum Zitat Zha S, Alt FW, Cheng HL, Brush JW, Li G. Defective DNA repair and increased genomic instability in Cernunnos-XLF-deficient murine ES cells. Proc Natl Acad Sci U S A. 2007;104(11):4518–23.PubMedPubMedCentralCrossRef Zha S, Alt FW, Cheng HL, Brush JW, Li G. Defective DNA repair and increased genomic instability in Cernunnos-XLF-deficient murine ES cells. Proc Natl Acad Sci U S A. 2007;104(11):4518–23.PubMedPubMedCentralCrossRef
78.
79.
Zurück zum Zitat Stewart DR, Pemov A, Johnston JJ, Sapp JC, Yeager M, He J, et al. Dubowitz syndrome is a complex comprised of multiple, genetically distinct and phenotypically overlapping disorders. PLoS One. 2014;9(6):e98686. Stewart DR, Pemov A, Johnston JJ, Sapp JC, Yeager M, He J, et al. Dubowitz syndrome is a complex comprised of multiple, genetically distinct and phenotypically overlapping disorders. PLoS One. 2014;9(6):e98686.
80.
Zurück zum Zitat Dasika GK, Lin SC, Zhao S, Sung P, Tomkinson A, Lee EY. DNA damage-induced cell cycle checkpoints and DNA strand break repair in development and tumorigenesis. Oncogene. 1999;18(55):7883–99.PubMedCrossRef Dasika GK, Lin SC, Zhao S, Sung P, Tomkinson A, Lee EY. DNA damage-induced cell cycle checkpoints and DNA strand break repair in development and tumorigenesis. Oncogene. 1999;18(55):7883–99.PubMedCrossRef
82.
Zurück zum Zitat Krempler A, Deckbar D, Jeggo PA, Lobrich M. An imperfect G2M checkpoint contributes to chromosome instability following irradiation of S and G2 phase cells. Cell Cycle. 2007;6(14):1682–6.PubMedCrossRef Krempler A, Deckbar D, Jeggo PA, Lobrich M. An imperfect G2M checkpoint contributes to chromosome instability following irradiation of S and G2 phase cells. Cell Cycle. 2007;6(14):1682–6.PubMedCrossRef
83.
Zurück zum Zitat Miglierina R, Le Coniat M, Gendron M, Berger R. Diagnosis of Fanconi’s anemia by flow cytometry. Nouv Rev Fr Hematol. 1990;32(6):391–3.PubMed Miglierina R, Le Coniat M, Gendron M, Berger R. Diagnosis of Fanconi’s anemia by flow cytometry. Nouv Rev Fr Hematol. 1990;32(6):391–3.PubMed
84.
Zurück zum Zitat Nahas SA, Butch AW, Du L, Gatti RA. Rapid flow cytometry-based structural maintenance of chromosomes 1 (SMC1) phosphorylation assay for identification of ataxia-telangiectasia homozygotes and heterozygotes. Clin Chem. 2009;55(3):463–72.PubMedPubMedCentralCrossRef Nahas SA, Butch AW, Du L, Gatti RA. Rapid flow cytometry-based structural maintenance of chromosomes 1 (SMC1) phosphorylation assay for identification of ataxia-telangiectasia homozygotes and heterozygotes. Clin Chem. 2009;55(3):463–72.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Rosen DB, Leung LY, Louie B, Cordeiro JA, Conroy A, Shapira I, et al. Quantitative measurement of alterations in DNA damage repair (DDR) pathways using single cell network profiling (SCNP). J Transl Med. 2014;12:184.PubMedPubMedCentralCrossRef Rosen DB, Leung LY, Louie B, Cordeiro JA, Conroy A, Shapira I, et al. Quantitative measurement of alterations in DNA damage repair (DDR) pathways using single cell network profiling (SCNP). J Transl Med. 2014;12:184.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Scully R, Xie A. Double strand break repair functions of histone H2AX. Mutat Res. 2013;750(1–2):5–14.PubMedCrossRef Scully R, Xie A. Double strand break repair functions of histone H2AX. Mutat Res. 2013;750(1–2):5–14.PubMedCrossRef
87.
Zurück zum Zitat Summers KC, Shen F, Sierra Potchanant EA, Phipps EA, Hickey RJ, Malkas LH. Phosphorylation: the molecular switch of double-strand break repair. Int J Proteomics. 2011;2011:373816. Summers KC, Shen F, Sierra Potchanant EA, Phipps EA, Hickey RJ, Malkas LH. Phosphorylation: the molecular switch of double-strand break repair. Int J Proteomics. 2011;2011:373816.
Metadaten
Titel
Biomarkers of DNA Damage Response Enable Flow Cytometry-Based Diagnostic to Identify Inborn DNA Repair Defects in Primary Immunodeficiencies
verfasst von
Kerstin Felgentreff
Ulrich Baumann
Christian Klemann
Catharina Schuetz
Dorothee Viemann
Martin Wetzke
Ulrich Pannicke
Sandra von Hardenberg
Bernd Auber
Klaus-Michael Debatin
Eva-Maria Jacobsen
Manfred Hoenig
Ansgar Schulz
Klaus Schwarz
Publikationsdatum
30.10.2021
Verlag
Springer US
Erschienen in
Journal of Clinical Immunology / Ausgabe 2/2022
Print ISSN: 0271-9142
Elektronische ISSN: 1573-2592
DOI
https://doi.org/10.1007/s10875-021-01156-7

Weitere Artikel der Ausgabe 2/2022

Journal of Clinical Immunology 2/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.