Skip to main content
Erschienen in: Acta Neuropathologica 4/2016

Open Access 01.04.2016 | Review

Mitochondria-associated membranes as hubs for neurodegeneration

verfasst von: Michiel Krols, Gert van Isterdael, Bob Asselbergh, Anna Kremer, Saskia Lippens, Vincent Timmerman, Sophie Janssens

Erschienen in: Acta Neuropathologica | Ausgabe 4/2016

Abstract

There is a growing appreciation that membrane-bound organelles in eukaryotic cells communicate directly with one another through direct membrane contact sites. Mitochondria-associated membranes are specialized subdomains of the endoplasmic reticulum that function as membrane contact sites between the endoplasmic reticulum and mitochondria. These sites have emerged as major players in lipid metabolism and calcium signaling. More recently also autophagy and mitochondrial dynamics have been found to be regulated at ER-mitochondria contact sites. Neurons critically depend on mitochondria-associated membranes as a means to exchange metabolites and signaling molecules between these organelles. This is underscored by the fact that genes affecting mitochondrial and endoplasmic reticulum homeostasis are clearly overrepresented in several hereditary neurodegenerative disorders. Conversely, the processes affected by the contact sites between the endoplasmic reticulum and mitochondria are widely implicated in neurodegeneration. This review will focus on the most recent data addressing the structural composition and function of the mitochondria-associated membranes. In addition, the 3D morphology of the contact sites as observed using volume electron microscopy is discussed. Finally, it will highlight the role of several key proteins associated with these contact sites that are involved not only in dementias, amyotrophic lateral sclerosis and Parkinson’s disease, but also in axonopathies such as hereditary spastic paraplegia and Charcot–Marie–Tooth disease.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00401-015-1528-7) contains supplementary material, which is available to authorized users.
Abkürzungen
AD
Alzheimer’s dementia
ALS
Amyotrophic lateral sclerosis
APP
Amyloid precursor protein
ATG
Autophagy-related
ATL1/3
Atlastin 1/3
CMT
Charcot–Marie–Tooth disease
CNX
Calnexin
DFCP1
Double FYVE-containing protein 1
DJ-1
Protein deglycase DJ-1
Drp1
Dynamin-related protein 1
EMC
Endoplasmic reticulum membrane complex
ER
Endoplasmic reticulum
ERMES
Endoplasmic reticulum-mitochondria encounter structure
FAD
Familial Alzheimer’s dementia
FIB-SEM
Focused ion beam scanning electron microscopy
Fis1
Fission 1 homolog
FTD
Frontotemporal dementia
GSK3β
Glycogen synthase kinase-3β
HSAN
Hereditary sensory and autonomous neuropathy
HSP
Hereditary spastic paraplegia
IMM
Inner mitochondrial membrane
INF2
Inverted formin 2
IP3R
Inositol 1,4,5-triphosphate receptor
MAM
Mitochondria-associated membrane
MCS
Membrane contact site
MCU
Mitochondrial calcium uniporter
Mff
Mitochondrial fission factor
MFN1/2
Mitofusin 1/2
miro1/2
Mitochondrial Rho GTPases 1/2
MT
Microtubule
OMM
Outer mitochondrial membrane
PA
Phosphatidic acid
PACS2
Phosphofurin acidic cluster sorting protein 2
PC
Phosphatidylcholine
PD
Parkinson’s disease
PE
Phosphatidylethanolamine
PEMT
Phosphatidylcholine-N-methyltransferase
PINK1
PTEN-induced putative kinase 1
PS1/2
Presenilin 1/2
PS
Phosphatidylserine
Pss1/2
Phosphatidylserine synthase 1/2
PTPIP51
Protein tyrosine phosphatase-interacting protein 51
REEP1/2
Receptor expression-enhancing protein 1/2
RTN2/4b
Reticulon 2/4b
RyR
Ryanodine receptor
SAD
Sporadic Alzheimer’s dementia
Sigma1R
Sigma non-opioid intracellular receptor 1
STX17
Syntaxin 17
TDP43
TAR DNA-binding protein 43
TEM
Transmission electron microscopy
Tom5
Outer mitochondrial membrane translocase complex 5
TRAK1/2
Trafficking kinesin protein 1/2
VAPB
Vesicle-associated membrane protein-associated protein B
VDAC1
Voltage-dependent anion channel 1
α-Syn
Alpha-synuclein

Mitochondria-associated membranes are specialized subdomains of the ER

Compartmentalization of biochemical reactions to dedicated membrane-bound organelles allows eukaryotic cells to perform the plethora of biological processes necessary to maintain homeostasis. While this has clear advantages, it also requires transfer of metabolites and signaling molecules between organelles to maintain cell performance. Such inter-organelle communication is often achieved via vesicular transport or transcriptional pathways. In recent years, alternative pathways involving direct communication between organelles through membrane contact sites (MCSs) have gained much interest. At MCSs, the membranes of two organelles are closely apposed through the formation of protein tethers, enabling fast, direct and reciprocal signaling between both compartments. With distances between the tip of the axon and the cell body reaching up to one meter in humans, the particular morphology of neurons poses limits on vesicular transport and transcriptional pathways. Processes that require strict regulation in space and time are therefore likely to depend much more on such MCSs. We focus this review on the contact sites that are formed between mitochondria and a specialized subdomain of the endoplasmic reticulum (ER) termed Mitochondria-Associated Membranes (MAMs). Emerging evidence shows that dysfunction of the MAMs plays a prominent role in numerous neurodegenerative diseases and that genes affecting ER and mitochondrial homeostasis are clearly overrepresented in hereditary disorders.
Close membrane appositions between the ER and mitochondria have been observed in electron micrographs as early as the 1950s [10, 24]. The ongoing development of subcellular fragmentation techniques repeatedly showed a resilient ER contamination in mitochondrial fractions, substantiating the existence of MCSs between the ER and mitochondria. Only in 1990, with the description of a membrane fraction ‘X’ associated with mitochondria and involved in phospholipid synthesis, the first function for this interface was identified [110]. This fraction ‘X’, now commonly referred to as the MAMs, is a specialized subdomain of the ER with a particular lipid and protein composition that is involved in the crosstalk with mitochondria. These ER-mitochondria contacts have since then been described in several organisms ranging from yeast to mammals [109]. Approximately 100 of such contacts are shown to occur in a single yeast cell and in mammalia approximately 5–20 % of the mitochondrial surface is estimated to be juxtaposed to the ER [1, 88]. Despite the ongoing development of superresolution techniques, the resolution of light microscopy is too limited for precise analysis of the morphology of ER-mitochondria contact sites, with the distance between both membranes at these sites being 10–30 nm. Transmission electron microscopy (TEM) does provide the required resolution; however, even when using tomography, it is very limited in the amount of 3D information which can be acquired. Volume electron microscopy techniques such as focused ion beam scanning electron microscopy (FIB-SEM) recently became available to obtain high-resolution 3D images of a whole cell and can thus be used as a powerful tool to gain more insight into the 3D morphology of MAMs [59]. Figure 1 and Supplementary movie 1 show the 3D reconstruction of mitochondria and their associated MAMs obtained using FIB-SEM imaging of a mouse embryonic fibroblast. These images clearly show that each mitochondrion makes multiple contacts with the ER and that all these contact sites are highly diverse in shape and size. It is currently unknown how MAM morphology relates to its function and therefore volume electron microscopy will be a crucial tool in studying the role different proteins play in regulating these contact sites.
Historically, the ER-mitochondria MCSs have been associated with phospholipid exchange between the ER and mitochondria [88]. In addition, the close proximity between both membranes appears to be critical for the efficient transfer of calcium from the ER to mitochondria. Further studies have revealed additional roles for ER-mitochondria MCSs in a variety of processes ranging from mitochondrial dynamics, sterol metabolism, autophagy, cell survival to energy metabolism and protein folding. Concomitant with the growing appreciation of this signaling hub, the list of disorders associated with ER-mitochondria MCSs is extending. To date this includes cancer [108], metabolic disorders such as Wolfram syndrome [114] or GM1-gangliosidosis [91], diabetes [104], viral infection [48], obesity [7], and neurodegeneration (see Table 1; Boxes 14).
Table 1
Overview of the genes affected in distinct neurodegenerative diseases and associated with MAM functions and/or structure
https://static-content.springer.com/image/art%3A10.1007%2Fs00401-015-1528-7/MediaObjects/401_2015_1528_Tab1_HTML.gif
Several observations indicate that mitochondria-ER contact sites play crucial roles in neuronal survival and death. Mitochondria in contact with the ER can be readily observed in brain tissue, throughout neurons and at synapses [47]. Already in 2002, it has been suggested that the choreographed interplay between the ER and mitochondria is involved in the shaping of dendritic calcium signals and neuronal activity of hippocampal neurons [84]. In addition at the synapse, calcium shuttling between both compartments was suggested to be essential in determining exocytosis and synaptic activity [74]. The involvement of signaling at the MAMs is implied in several neurodegenerative disorders including Alzheimer’s dementia (AD) [93], Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD) [9, 85, 97], Parkinson’s Disease (PD) [17, 42, 82] and Charcot–Marie–Tooth disease (CMT) [28] (for a comprehensive overview see Table 1; Boxes 14). More generally, many if not all processes linked to ER-mitochondria contacts are widely implied in neurodegenerative disorders. This review will focus on the structure and function of MAMs and how their disruption contributes to neurodegeneration.

Structural composition of the MAMs

The MAM fraction can be detached from mitochondria through proteolysis with trypsin or Proteinase K, showing that a proteinaceous tether is responsible for connecting both membranes [26]. The presence of such a protein bridge has been confirmed in electron microscopy studies [26, 97], but despite this, the identity of its components still remains largely elusive. Most proteins that have been found to be enriched at the MAMs, are unlikely to participate directly in tethering, but might be functionally involved in the processes mediated by these contacts [108, 109].
In yeast, genetic screens independently led to the discovery of the ER-Mitochondria Encounter Structure (ERMES) complex [55] and the ER membrane protein complex (EMC) [60] as functional tethers between the ER and mitochondria (see Fig. 2 lower part). The ERMES complex consists of the outer mitochondrial membrane proteins Mdm10 and Mdm34, the ER membrane protein Mmm1 and the cytosolic Mdm12. Loss of any of these proteins could be rescued by the expression of an artificial linker, establishing the structural role of ERMES. Gem1, the yeast ortholog of the mitochondrial Rho GTPases 1 and 2 (Miro1 and Miro2), and more recently also Lam6, were identified as regulators of the ERMES complex [32, 56, 78]. A second screen identified the interaction between the EMC and the outer mitochondrial membrane (OMM) translocase complex 5 (Tom5) as an interaction bridging the ER and mitochondrial membranes independent of ERMES (Fig. 2 lower part). In contrast to the ERMES complex, this contact is essential in the lipid exchange between both organelles [60]. While Lam6, Gem1 and the EMC proteins do have metazoan orthologs, it is as yet unclear whether these orthologs effectively function as regulators of interorganelle contact in mammalia.
The first complex identified in mammalian cells that bridges the gap between the MAM and the OMM is the tripartite complex between the cytosolic chaperone Grp75, the mitochondrial voltage-dependent anion channel 1 (VDAC1) and the inositol 1,4,5-triphosphate receptor (IP3R) in the MAM [100] (Fig. 2 upper part). While knockdown of Grp75 in HeLa cells decreases the transfer of calcium from the ER to mitochondria, ER-mitochondrial linkage is unaffected in cells lacking all three IP3R isoforms [26]. Therefore, rather than representing a structural tether, this interaction is considered a functional one, promoting efficient transfer of calcium from the ER to mitochondria (see below). Other candidate protein tethers are the homodimers or heterodimers of ER-resident Mitofusin (MFN) 2, a dynamin-related GTPase, with MFN1 or MFN2 on the OMM (Fig. 2 upper part) [28]. Cells depleted of MFN2 were reported to have decreased ER-mitochondria contact and crosstalk [5, 28, 44, 82]. CMT-causing mutations in MFN2 failed to rescue the disrupted ER-mitochondria tethering (see Box 4; Table 1) [28]. MFN2 dependent ER-mitochondria crosstalk is tightly controlled by ubiquitination, further supporting its essential role [98]. Still, independent studies could not confirm the crosstalk-promoting role of MFN2 in ER-mitochondria contacts, but rather reported the opposite finding, i.e., loss of MFN2 led to an increase in MCS [25, 34]. These contradictory findings remain to be resolved, as does the effect of CMT-causing mutations in MFN2 on MAM signaling (see Table 1; Box 4).
Adding to this complexity, additional tethering interactions have been identified, which might compensate for MFN2 loss in certain conditions. One such tether is the physical interaction between the MAM protein VAMP (vesicle-associated membrane protein)-associated protein B and C (VAPB) and the mitochondrial protein tyrosine phosphatase-interacting protein 51 (PTPIP51) (Fig. 2 upper part) [29, 97]. Overexpression of either or both of these proteins increases the extent of contact while loss of either protein through siRNA-mediated knockdown significantly diminishes membrane contact and crosstalk [97]. A functional rather than a structural interaction between the ER and mitochondria has been uncovered by establishing the interaction between ER-resident BAP31 and the mitochondrial fission protein Fission 1 homolog (Fis1) [50]. The complex formed between BAP31/Fis1 constitutes an apoptosis-signaling platform needed for the recruitment of procaspase 8, the cleavage of BAP31 and a subsequent increase in Ca2+ release from the ER to the mitochondria [50]. This fits with earlier notions that a feedback ER contribution is needed to amplify mitochondrial apoptosis signals [3]. Finally, the vesicular sorting protein phosphofurin acidic cluster sorting protein 2 (PACS2) contributes to the structural integrity of the ER-mitochondria MCSs, through a stabilization mechanism that is not entirely clear [95] (Fig. 2 upper part). The siRNA-mediated knockdown of PACS2 uncouples the ER from mitochondria and induces BAP31 cleavage-dependent mitochondrial fragmentation and cell death in A7 melanoma cells [95], and similarly results in caspase-3 mediated degeneration of primary hippocampal neurons and astrocytes [47]. The complex role of PACS2 as a modulator of MAM properties is discussed further.
Although the mediators of these MCSs seem to be quite distinct between yeast and mammals, the property that different and independent linkers are involved in the coupling of both membranes is conserved. The relevance of having diverse connections is currently unclear, however, it does suggest that these could serve distinct functions. A recent study in mammalian cells indeed indicates the presence of two distinct domains in MAMs that differ in their lipid content and function [4], yet more research is needed to shed light on how distinct protein and lipid compositions can affect MAM function.

Several essential cellular processes are controlled by MAMs

As stated in the introduction, recent studies revealed several novel functions for the MAMs. In the following sections we will give a brief description of the most important MAM functions in the context of neurobiology.

Regulation of lipid metabolism

Although most membrane lipid synthesis in eukaryotic cells occurs in the ER, several key metabolic steps and lipid modifications are performed in other organelles, and thus, require transport of lipids. How hydrophobic lipids are distributed within the aqueous environment of the cell towards other organelles remains largely elusive, but recently the role of non-vesicular transport at membrane contact sites has gained a lot of interest [61, 109]. Close membrane apposition at these sites appears to allow efficient shuttling of lipids. In addition, several lipid metabolic enzymes are exclusively localized at certain MCSs, restricting the generation of these lipid species to the site where they are needed. MAMs were originally identified as a fraction highly enriched in phosphatidylserine (PS) synthase-1 and -2 (Pss1 and Pss2) and transfer of phospholipids between the ER and mitochondria was the first function ascribed to ER-mitochondria contacts [109]. Phosphatidylserine, newly synthesized in the MAM by the PS synthases, is subsequently transferred to the closely apposed mitochondrion (Fig. 3). In the inner mitochondrial membrane (IMM), decarboxylation of PS results in the production of phosphatidylethanolamine (PE). The importance of ER-mitochondria MCSs is underlined by the fact that the transfer of PS between both organelles is the rate-limiting step in PE synthesis from PS [112]. In a crude mitochondrial fraction derived from rat liver, newly synthesized PE can then be transferred back to the MAM where it is converted into phosphatidylcholine (PC) by the enzyme PE-N-methyltransferase (PEMT) [110]. Since PEMT activity is restricted to hepatocytes, it is not clear how essential MAMs are for PC synthesis in other tissues.
Protein identification approaches revealed that not only Pss1 and Pss2 are enriched at the MAMs, but also several other lipid synthesizing enzymes involved in cholesterol and sphingolipid biosynthesis (Fig. 3) [39, 45, 109]. This fits well with thin layer chromatography studies showing that MAMs comprise a unique lipid profile, with highly elevated levels of ceramides and cholesterol compared to the bulk ER [45]. While it is yet unclear whether the MAMs participate in trafficking of these lipids to mitochondria, this particular lipid raft-like composition plays a determining factor in the recruitment of several well-established MAM components. Both presenilin 1 and 2 (PS1 and PS2), members of the γ-secretase complex are targeted to the MAMs because of their lipid raft composition [106]. Similarly, the chaperone Sigma non-opioid intracellular receptor 1 (Sigma1R), associated with AD, ALS and FTD is strictly dependent on the presence of cholesterol and ceramide for targeting to the MAMs [45] and for performing its essential functions in calcium regulation (see Table 1; Fig. 4).

Regulation of calcium homeostasis at the MAMs

Already in the 19th century, the role of calcium as second messenger was established through seminal experiments on perfusion of isolated hearts. Since then, a lot of research has been devoted to understand how increases in calcium levels (so-called oscillations) are generated, decoded and translated in signaling events. Two major sources count for the rise in cytosolic calcium: the extracellular medium with a [Ca2+] of ~1 mM and the ER with a [Ca2+] >100 μM [87]. Mitochondria play a crucial role in buffering increases in cytosolic calcium, and a rise in cytosolic [Ca2+]cyt is always closely followed by a rise in [Ca2+]mito [89]. Interestingly, upon ER calcium release, mitochondrial levels can rapidly increase 10–100-fold without noticeable rises of cytosolic calcium levels [87]. In contrast to the outer mitochondrial membrane (OMM), which is permeable to calcium ions, influx of calcium into the mitochondrial matrix—driven by the electrical gradient across the IMM—occurs through the recently identified mitochondrial calcium uniporter (MCU) complex. This complex consists of the MCU protein and its regulators (MICU1, MICU2, MCUb, MCUR1, and EMRE) [72]. Paradoxically, the low calcium affinity of the MCU appears incompatible with the observed rapid calcium uptake in mitochondria at low cytosolic calcium levels. This discrepancy was solved by the observation that mitochondria are in close contact with the IP3Rs and ryanodine sensitive channels (RyRs) mediating ER store release [88]. This occurs at the MAMs and leads to the transient formation of “hotspots”, perimitochondrial microdomains in which calcium levels far exceed those in the bulk of the cytosol, enabling rapid import across the IMM [27, 40, 88].
ER-mitochondria MCSs are able to establish such microdomains required for efficient calcium transfer through (1) the close apposition of both membranes, (2) the enrichment of the calcium release channels IP3Rs and RyRs at MAMs and (3) the connection of IP3Rs to the mitochondrial VDAC1 by the chaperone Grp75 (Fig. 4). Knockdown of Grp75 abolishes functional coupling between VDAC1 and IP3R, resulting in diminished calcium transfer [100]. On the other hand, bringing the ER closer to mitochondria through an artificial linker stimulates calcium transfer [26]. Numerous studies have correlated altered ER-mitochondrial connectivity to alterations in calcium transfer between both organelles, through overexpression or knockdown of proposed tethers, including MFN2 and VAPB-PTPIP51, or regulators thereof [28, 29, 34, 97] (Fig. 4). Several modulators of IP3R channel activity are present at or recruited to MAMs under various conditions. For example, various ER-resident chaperones and oxidoreductases can affect the exchange of calcium between the ER and mitochondria [96]. Calnexin (CNX) modulates local calcium levels by acting as a calcium buffer and through regulating the activity of IP3R and the sarco/endoplasmic reticulum calcium ATPase (Serca), the main ATPase pumping cytosolic calcium into the ER [52, 68, 90]. CNX targeting to the MAM is dependent on palmitoylation [68], and on the cytosolic sorting protein PACS2 [79] and Rab32 [16] two trafficking molecules that both regulate the composition of the MAMs (Fig. 4). The ER-resident Sigma1R is released from Bip upon ER calcium release, and binds to IP3Rs at the MAMs, in this way preventing their proteasomal degradation and sustaining prolonged Ca2+ uptake by mitochondria [46] (Fig. 4). Especially in neurons, both Sigma1R and PACS2 play a crucial role in mediating calcium homeostasis and loss of Sigma1R results in neuromuscular defects [47]. This is underscored by the fact that alterations in PACS2 and Sigma1R levels are associated with the pathogenesis of AD, ALS and FTD (see Table 1; Boxes 1, 2). In addition, presenilin mutations in AD are associated with an increased IP3R calcium release (see Table 1; Box 1). Finally, in Huntington’s disease ER-mitochondrial calcium transfer appears affected as well. Mutant huntingtin, but not the wild type protein, was found to interact with IP3R and facilitate calcium transfer between the ER and mitochondria [12, 83].
Calcium transfer to mitochondria occurs under both physiological and stress conditions. A moderate mitochondrial matrix calcium increase stimulates ATP synthesis through calcium dependency of metabolic enzymes in the Krebs cycle [87]. Functional coupling of the ER and mitochondria therefore stimulates aerobic metabolism, in response to a demand of ATP-requiring processes in the cytosol as well as in the ER [96, 111]. Hence, loss of contact has adverse effects on ER homeostasis, protein folding and energy metabolism [14], and will affect cell survival and proliferation, especially in cells with high-energy demands such as neurons. Mitochondrial calcium overload on the other hand is a potent inducer of the mitochondrial permeability transition pore and can thereby lead to OMM rupture and the escape of pro-apoptotic factors such as cytochrome c into the cytosol [87]. In this respect, regulation of ER-mitochondrial calcium transfer at the MAMs is essential in the switch between cell survival and death during various stress responses. In the context of Huntington’s disease for example, the binding of mutant huntingtin to IP3R may result in a heightened susceptibility of mitochondria to activate the permeability transition pore and the subsequent release of pro-apoptotic factors [23]. Intriguingly, proteins that modulate calcium transfer between mitochondria and the ER are affected in many neurodegenerative diseases and could be tightly linked to a loss in neuronal survival [94] (see Table 1).

Regulation of mitochondrial dynamics and homeostasis

The function of both the ER and mitochondria strongly depends on a highly regulated balance in membrane dynamics. Mitochondrial dynamics include their transport along cytoskeletal tracks throughout the cell, in addition to regulated fission and fusion. Combined, these dynamics ensure optimal mitochondrial metabolism and, in neurons, the correct distribution of mitochondria to dendrites and synapses. Especially in neurons with long axons this poses a challenge, which is highlighted by the many diseases caused by defects in the proteins involved in mitochondrial dynamics [19] (see Boxes 24; Table 1). Mitochondrial fission is known to require the assembly of dynamin-related protein 1 (Drp1) in a helix around the OMM, followed by constriction of this ring (Fig. 5). However, it has long been unclear what signaling events precede this process and mark the site of Drp1 assembly. In 2011, Friedman et al. showed in Cos7 cells that focal accumulation of Drp1 and subsequent fission occurs at or near sites of contact with the ER, and the same holds true for the yeast Drp1 ortholog Dnm1 [37]. Mitochondrial constriction occurs in the vicinity of the ER even in the absence of Drp1 and the mitochondrial fission factor (Mff). These observations suggested that ER tubules mark the sites of mitochondrial fission and induce initial constriction, followed by the recruitment of Mff and finally Drp1 helix assembly that mediates the division [37]. Indeed, recent studies have described that actin polymerization, induced at the MCSs by an ER-bound isoform of Inverted Formin 2 (INF2), is capable and required to induce the initial constriction required for mitochondrial fission (Fig. 5) [58]. Spire1C, a newly described mitochondrial isoform of the actin-nucleating Spire proteins, interacts with INF2 to specifically nucleate actin filaments only at sites where the ER and mitochondrial membranes are closely apposed. Loss of this interaction results in long tubular mitochondria reminiscent of defective fission [70]. Mitochondrial constriction prior to Drp1 assembly depends on this actin polymerization step, however, additional force generated by Myosin II may also contribute to this process [30, 57]. How mutations in INF2, causing Charcot–Marie–Tooth disease type 2D and glomerulopathy [15] affect this process remains to be determined. Finally, it was shown in a very elegant way that Drp1 activity at the MAMs is strictly regulated by phosphorylation in a process that involves Rab32 and syntaxin 17 (STX17) [4] (Fig. 5). This process seems tightly linked to the regulation of calcium homeostasis and the initiation of autophagy (see further). In addition, SUMOylation of Drp1 was reported to affect its function at the MAM, stabilizing the ER-mitochondria contact site and thus promoting calcium crosstalk and cytochrome c release [86].
In contrast to the role of Gem1p in ER-associated mitochondrial division in yeast, no such role has been described for the mammalian Gem1 orthologs Miro1 and Miro2. In mammalian cells, both Miro proteins 1 and 2 are known as essential regulators of mitochondrial motility [36, 69, 92]. These atypical Rho GTPases are anchored in the OMM and mediate anterograde and retrograde transport by connecting the mitochondria to microtubule (MT)-bound kinesin and dynein through the trafficking kinesin (TRAK1 and TRAK2) adaptor proteins (Fig. 5) [36, 69]. Neuron-specific loss of Miro1 in mice results in a neuronal disease phenotype that most closely resembles that of spastic paraplegia patients [80] (see Box 4; Table 1).
Interestingly, several observations point to a role of MAMs in Miro1/2-mediated cellular transport of mitochondria. First, as mentioned, the yeast ortholog of Miro1/2 is a regulatory subunit of the ERMES complex and Miro1 localizes at ER-mitochondria contact sites in mammalian cells [56]. Second, at sites of close contact, the ER and mitochondria move along acetylated MTs while staying attached, suggesting coordinated transport [38]. Third, in addition to coordinating fusion and tethering to the ER of the OMM, MFN2 is required for axonal transport and both MFN1 and MFN2 interact with Miro1/2 [75]. Fourth, mitochondrial transport is regulated by calcium through the EF-hand motifs in Miro1/2, which function as a calcium sensor. Calcium binding of Miro1/2 induces a conformational change that disconnects the mitochondria from the MT track it moves along, thus halting its transport [20, 69, 92]. Due to the relatively low affinity of Miro1/2 to calcium, calcium-dependent halting of mitochondrial transport is most likely to occur at calcium hotspots, such as the ER-mitochondria MCSs [20, 92]. The dependence on high calcium regions indicates a mechanism whereby the proximity with the ER can determine the redistribution of mitochondria. This provides the cell with a powerful system of targeting mitochondria based on local energy requirements. Particularly in neurons, this might be a relevant factor in determining the distribution of mitochondria to the dendrites, synapses and the nodes of Ranvier that is currently insufficiently understood. That such a system is essential for neuronal survival is underscored by the many reports of mitochondrial transport defects in neurodegenerative disorders [19]. Much research has focused on the connection between mitochondria and MTs in this context. It is becoming increasingly clear, however, that the connection with the ER is also a major determinant for axonal transport of mitochondria (see Boxes 24).

MAMs as a potential regulator of autophagy and mitophagy

Macroautophagy is the process whereby damaged proteins and organelles are cleared from the cell by sequestering them in a double membrane-bound vesicle termed the autophagosome. Subsequent delivery to the lysosome allows proteasomal breakdown and recycling of the substrates. Debate is still ongoing with respect to the origin of the phagophore and the identity of membrane donor sources needed for its expansion to form the mature autophagosome [62]. Axe et al. identified the omegasome, a phosphatidylinositol-3-phosphate-enriched membrane structure attached to the ER, as a membrane source for phagophore development [8]. In addition, the mitochondria, harboring many autophagy-related (ATG) proteins and regulators thereof, have been proposed as an origin for phagophore formation, based on the transfer of a fluorescently labeled mitochondrial marker to the phagosome [43]. Intriguingly, several ATG proteins have been found to accumulate specifically at ER-mitochondria MCSs in conditions of starvation, and more and more evidence is accumulating that MAMs might be the actual site of autophagosome formation, unifying both earlier models [43]. Hailey et al. first speculated on the requirement of lipid transfer from the ER to mitochondria during autophagosome biogenesis. In mouse cells lacking MFN2, autophagy induction was disturbed. The authors contributed this to a decreased lipid transfer towards mitochondria as a consequence of ER-mitochondrial uncoupling and concluded that lipids transferred from the ER accumulate in the OMM, from where they are trafficked to the expanding phagophore [43] (Fig. 6). A more direct involvement of MAMs in autophagosome biogenesis was proposed by Hamasaki and coworkers [44]. Upon starvation of mammalian cells, Vps15, Vps34, Atg14L, Beclin1—early markers of phagophore formation—and the omegasome marker double FYVE-containing protein 1 (DFCP1) accumulate in the MAM fraction (Fig. 6) [44]. This might be initiated by early translocation of STX17 [4]. In starvation conditions, STX17 translocates in an mTOR/ULK dependent manner to a domain of ER-mitochondrial contact resistant to digitonin, where it recruits ATG14L [4]. ATG14L then interacts with the phosphoinositide 3-kinase complex consisting of Beclin1, p150 and Vps34, resulting in local phosphatidylinositol-triphosphate production. This alters the local lipid composition of the MAM membrane, which leads to the recruitment of WD-repeat domain phosphoinositide interacting (WIPI) proteins, the effector proteins and the mediators of autophagosome biogenesis. It has been proposed that this could be sufficient to induce a deformation of the membrane [62]. Consistent with a role for the MAMs in autophagosome formation, knockdown of MFN2 or PACS2 abolished ATG14L puncta formation and downstream LC3 lipidation in starved cells, and caused defective STX17 localization at the MAM [44].
In addition, during the selective autophagy of mitochondria, known as mitophagy, ER-mitochondria contact sites appear to constitute a platform promoting mitophagosome formation. In yeast, efficient mitophagy depends on coupling of mitochondria and the ER through ERMES, while bulk autophagy can apparently operate in the absence of this tether [13]. ERMES colocalizes with sites of mitophagosome biogenesis and affects the formation of the phagophore that engulfs the mitochondria destined for degradation [13, 71].
In mammalian cells, mitochondria destined for degradation recruit the E3 ubiquitin ligase Parkin to the OMM through PTEN-induced putative kinase 1 (PINK1) kinase activity. Ubiquitination of Parkin substrates subsequently recruits the autophagy machinery [31]. ER-mitochondria MCSs are implied in this process in several ways. First, focal accumulation of DFCP1 along mitochondria labeled for degradation with exogenously expressed Parkin suggests a role for the ER/MAM as a membrane source for the mitophagosome [115]. Second, Drp1-mediated fission, at least in part mediated by contacts with the ER [37], is suggested to promote mitophagy by breaking off pieces of mitochondria promoting their engulfment by the mitophagosome [105]. Lastly, depolarization of the IMM through calcium (over)loading could be involved in PINK1 and Parkin translocation to the OMM, activating mitophagy [31, 53].
While the exact mechanisms governing autophagy induction and progression are not entirely clear, several lines of evidence point to an essential role of the ER-mitochondria interface during this process. The presence at the MAMs of several important players in metabolism, such as mTORC2 [11], allows to speculate that ER-mitochondria MCSs could represent a site of crosstalk between sensors of the cells’ energetic needs and the early players in autophagy. Additionally, through interorganelle communication, dysfunctional mitochondria may be sensed at the MAMs, followed by mitophagy induction and targeted removal of these unhealthy mitochondria. Future research is needed to study these mechanisms and how they are disrupted in disease. Dysregulation of neuronal energy metabolism and mitochondrial homeostasis are important factors contributing to neurodegeneration [94]. This appears relevant in particular for PD (see Box 3), however, autophagy is a process whose dysregulation is widely implied in neurodegenerative disorders [73].

Conclusion

MCSs provide pathways of intracellular signaling that are only beginning to be unraveled. Although many questions remain as to how ER-mitochondria MCSs are maintained and regulated, it is clear that many different pathways intertwine at these signaling hubs. Whether all the different functions described here occur simultaneously at the same MCS, or whether specialized ER-mitochondria MCSs exist that mediate a subset of these processes is currently unclear. Gaining further insight into the protein tethers that connect both organelles will be crucial to shed more light on the regulation of MAM functions.
As these functions become uncovered, also the relevance of ER-mitochondria contact sites in disease becomes apparent. Neuronal cells in particular appear to be very vulnerable to insults affecting the balance in ER-mitochondrial communication. Indeed, many of the processes that require these contact sites are implied in neurodegeneration. As such, resolving the role of MAMs in the pathomechanisms leading to these disorders may unify several of the previously identified defects that occur during neurodegeneration, including imbalances in calcium or lipid signaling, mitochondrial dynamics and autophagy pathways. Since tipping the balance in either direction appears detrimental for neuronal survival, uncovering the different players that maintain this delicate equilibrium will be crucial to understand the role that MCSs play in these disorders and possibly lead to novel strategies to reverse ER-mitochondrial homeostasis defects in patients.

Box 1: Alzheimers’s dementia

Aberrant cleavage of the amyloid precursor protein (APP) is the main hallmark of AD pathogenesis. Mutations in PS1 and PS2, components of the ɣ-secretase complex involved in APP processing, are major causes of familial AD (FAD) (Table 1). Consistent with their known association with lipid rafts, PS1, PS2, APP and ɣ-secretase activity are enriched at MAMs [6]. Moreover, in cells deficient of MFN2, which showed diminished MAM function in terms of lipid metabolism, the enzymatic activity of the ɣ-secretase was reduced by approximately 50 %, showing that APP processing relies on intact ER-mitochondria crosstalk [5]. Vice versa, MAM function also depends on PS1 and PS2. ER-mitochondria MCSs and MAM-dependent lipid metabolism were shown to be upregulated in cells deficient of PS1 and PS2 as well as in cell models of both familial and sporadic AD (SAD) [5]. The complicated interplay between the ɣ-secretase constituents and activity and ER-mitochondrial crosstalk also extends to calcium signaling. The PS1 and PS2 proteins were shown to interact with IP3Rs and FAD-associated mutations in either gene result in a higher IP3R gating activity and exaggerated calcium release from the ER, followed by increased APP cleavage [21, 22]. In turn, exposing hippocampal neurons or neuroblastoma cells to amyloid β enhances ER-mitochondria contact and calcium transfer [33, 47]. Post-mortem analysis of human AD brain and those of AD mouse models showed altered expression levels of PACS2 and Sigma1R, MAM proteins involved in calcium handling, thus further establishing dysregulated MAM signaling as a hallmark of AD [47]. How exactly MAM function influences APP processing and how a defect in the ɣ-secretase in turn affects ER-mitochondria MCSs remains to be further clarified. The finding that MAMs are involved early in the pathogenesis of both FAD and SAD is a major breakthrough nonetheless, as it can explain many of the different neuronal defects associated with AD, including calcium homeostasis, mitochondrial dysfunction, oxidative stress and lipid metabolism [93]. Most recently, it was also reported that the E4 allele of the apolipoprotein E (ApoE4), a major risk factor for developing SAD, contributes to increased crosstalk at the ER-mitochondria MCS [101]. MAMs are thus placed center-stage in the AD pathogenesis and therefore make for a promising target for therapeutics. One such target could be GSK3β, a central regulator of homeostasis in the brain that is implied in a large number of disorders including AD [66]. As further elaborated upon in BOX 2, GSK3β was recently identified as a negative regulator of ER-mitochondria MCSs formation [97].

Box 2: Amyotrophic lateral sclerosis

MAM integrity and signaling have been found affected in several ALS subtypes. VAPB is an ER protein involved in tethering the MAM to mitochondria [29]. A point mutation in the gene encoding VAPB was identified in patients suffering from the severe familial ALS type 8 [81] (Table 1). Mutant VAPB is enriched at the MAM, where increased binding with PTPIP51 tightens the connection at the ER-mitochondria interface and results in higher mitochondrial calcium peaks (Fig. 4) [29]. More recently, two other proteins involved in ALS have been identified as modulators of this interaction: glycogen synthase kinase-3β (GSK3β), established as a negative regulator of VAPB/PTPIP51 interactions, and TAR DNA-binding protein 43 (TDP43), which activates GSK3β [97] (Figs. 2, 4). GSK3β hyperactivity is not only connected to ALS, but it also takes a central role in the disease mechanisms leading up to AD and PD, both in familial and sporadic cases [41, 66] (Table 1). TDP43 accumulation on the other hand is a hallmark of the FTD/ALS pathology and mutations in the TDP43 gene occur in approximately 3 % of patients with familial ALS as well as in 1.5 % of sporadic cases [63] (Table 1). Overexpression of TDP43 leads to an activation of GSK-3β and consequently weakens ER-mitochondria coupling [97]. In a transgenic TDP43 mouse model for ALS, motor neurons show reduced contact between the ER and mitochondria. Similarly, overexpression of wild type or mutant TDP43 in NSC34 cells disrupts both the structural and the functional connection between both organelles, ensued by a diminished exchange of calcium [97]. The fact that in one ALS subtype calcium signaling at MAMs is found to be upregulated, whereas it appears downregulated in another, underscores the importance of balanced communication between both organelles for neuronal survival.
In addition to tightened contact between the ER and mitochondria and elevated calcium transfer, expression of VAPB P56S disrupts the connection of Miro1 to tubulin, obstructing anterograde, but not retrograde mitochondrial transport in rat cortical neurons [77]. This phenotype was rescued by the co-expression of a calcium-insensitive Miro1 mutant, emphasizing the importance of controlled calcium signaling at MAMs for mitochondrial transport [77]. Of note, an earlier report showed that fibroblasts derived from VAPB P56S patients as well as neuronal cultures displayed intracellular aggregates containing VAPB and Sigma1R, a major regulator of calcium trafficking at MAMs [46]. Moreover, pharmacological Sigma1R activation was neuroprotective in this context and led to decreased VAPB aggregation [85]. Interestingly, mutations in the gene coding for Sigma1R are also associated with ALS/FTD [2, 67]. A physiological connection between VAPB and Sigma1R at the MAMs remains to be determined, however, the defect in mitochondrial dynamics upon loss of Sigma1R points to a shared function of both proteins in ER-regulated axonal transport of mitochondria [9]. As the large majority of ALS cases are sporadic rather than familial, it will be crucial to study whether MAMs also play a role in these sporadic cases.

Box 3: Parkinson’s disease

Parkinson’s disease is caused by loss of dopaminergic neurons in the substantia nigra. A minority of PD cases are monogenic, caused by mutations in the genes encoding Parkin, PINK1, alpha-Synuclein (α-Syn) or the protein deglycase DJ-1 amongst others [94]. Parkin and PINK1, as mentioned, play important roles in mitophagy and mutations in the genes encoding these proteins are believed to disrupt the proper degradation of defective mitochondria [31]. The function of Parkin, however, appears broader than merely marking damaged mitochondria. In cultured rat neurons, glutamate excitotoxicity elicits the accumulation of Parkin at mitochondria, the ER and the ER-mitochondrial interface, without inducing mitophagy [107]. Overexpression of Parkin was previously shown to increase physical and functional coupling of the ER and mitochondria, stimulating ATP production and calcium exchange, while knockdown has the opposite effect and disrupts mitochondrial morphology [17]. The exact function of Parkin at the ER or its junctions with mitochondria and whether and how this is connected to its function in mitophagy is currently unclear, however, these observations point to a protective role for Parkin during excitotoxic stress, by maintaining ER-mitochondrial crosstalk.
Like Parkin, also α-Syn and DJ-1 overexpression promotes MAM function and interaction with mitochondria (Fig. 2) [18, 42, 82]. Intriguingly, all these proteins were reported to interact with Grp75, providing a plausible mechanism whereby they can affect ER-mitochondria calcium signaling, although alternative pathways may be involved as well [51]. Wild type α-Syn, known to have a high affinity for lipid rafts, localizes to MAMs where it promotes physical contact with mitochondria [18]. Consistent with a role for α-Syn in contact formation, loss of α-Syn at the MAMs results in diminished ER-mitochondria signaling [18, 42]. In contrast to wild type α-Syn, PD-causing mutant forms of α-Syn downregulate ER-mitochondrial apposition, presumably through disrupted interaction with lipid rafts in the case of A30P mutant α-Syn, or through decreased levels of total α-Syn in the case of the A53T mutant [5, 35]. Accumulation of α-Syn in protein aggregates termed Lewy bodies, a major hallmark of both familial and sporadic PD, could therefore represent of loss of function of this protein at the MAMs, which may underlie the disorder.
No direct involvement for PINK1 in ER-mitochondrial crosstalk has been reported. It does, however, play a role in mitochondrial motility, regulation of which also takes place at this interface. In addition to local calcium levels, mitochondrial motility is regulated through proteasomal breakdown of the transport complex, as PINK1 promotes the proteasomal degradation of Miro1/2 and MFN1/2 in a Parkin-dependent manner [65, 102, 113]. Intriguingly, loss of Miro was shown to rescue the phenotype caused by expression of PD-causing PINK1 mutations in Drosophila and facilitates clearance of damaged mitochondria in HeLa cells, suggesting that this process plays a significant role in the disease [65]. Further research is needed to investigate how contacts of mitochondria with the ER are involved in this pathway and whether ubiquitination of MFN2 by Parkin can modulate these contacts, as reported for MITOL [98]. Combined, these findings accentuate the complex interdependence of mitochondrial quality control and mitochondrial dynamics, pathways that clearly intersect at the ER-mitochondrial interface.

Box 4: Axonopathies

CMT and HSP are both caused by degeneration of long axons, in the case of CMT those of the peripheral motor and/or sensory nerves, while the upper motor neurons are predominantly affected in the case of HSP. Unsurprisingly, these disorders share many commonalities with regards to the pathomechanisms involved and a disproportionate number of affected genes localizes to mitochondria, the ER, or the MCSs shared by both [103]. In HSP, membrane shaping of the ER is a major theme, with involved genes including Receptor Expression-Enhancing Protein 1 and 2 (REEP1/2), Spastin, Reticulon 2 (RTN2) and Atlastin 1 (ATL1). ATL1 and ATL3 are also found mutated in hereditary sensory and autonomous neuropathy (HSAN) [49, 54]. Work in yeast suggests that defects in ER-shaping proteins may affect ER-mitochondrial MCSs and lipid transfer. Interestingly, the RTN2 homolog RTN4b was identified as a negative regulator of crosstalk between both organelles [99]. More recently, HSP-causing mutations in or loss of REEP1 were also reported to disrupt ER-mitochondria MCSs [64]. Though no direct effect on MCSs was reported, neurons differentiated from patient-derived induced pluripotent stem cells carrying the HSP-causing P342S mutation in ATL1 display not only defects in ER morphology, but also in the axonal transport of mitochondria [116]. Another example of an ER-bound protein that modulates mitochondrial dynamics is INF2, mutations in which cause CMT associated with glomerulopathy [15]. INF2 modulates actin dynamics required for MAM-dependent mitochondrial fission (see main text and Fig. 5) [58, 70]. How CMT-causing mutations in this gene affect mitochondrial dynamics remains to be elucidated, however, it is clear that ER-resident proteins can play significant roles in mitochondrial dynamics through the interface between both organelles. Similarly, proteins known for their role in mitochondrial dynamics can affect the ER network. CMT-causing mutations in MFN2 disrupt ER morphology and contact with mitochondria [28], in addition to interfering with mitochondrial dynamics and distribution within axons [75, 76]. Since these mutations do not affect the interaction of MFN2 with Miro1/2, aberrant connectivity with the ER might be the underlying factor causing mislocalization of mitochondria within axons [76]. It is clear that the interplay between ER and mitochondrial network dynamics and interorganelle communication plays a pivotal role in axonal survival and degeneration. Though the exact mechanisms controlling mitochondrial motility at the MCSs with the ER are the subject of ongoing research, several observations suggest that a multitude of pathways can impinge on this process and it is likely that the signaling hub where these pathways are integrated, are the ER-mitochondria contact sites.

Acknowledgments

M. K. is supported by a PhD fellowship of the Fund for Scientific Research (FWO-Flanders, Belgium). The research of V.T. is supported by the FWO, University of Antwerp, the Association Belge contre les Maladies Neuro-Musculaires (ABMM) and the EU FP7/2007–2013 under Grant Agreement Number 2012-305121 (NEUROMICS). The research of S. J. is supported by the FWO, University of Ghent and GROUP-ID. The Zeiss Auriga was acquired through a clem grant from Minister Ingrid Lieten to the VIB Bio Imaging Core. Special thanks to Sonia Bartunkova for technical support and Benjamin Pavie for advise in image processing.
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.

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 Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

e.Med Neurologie

Kombi-Abonnement

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

Literatur
1.
Zurück zum Zitat Achleitner G, Gaigg B, Krasser A, Kainersdorfer E, Kohlwein SD, Perktold A, Zellnig G, Daum G (1999) Association between the endoplasmic reticulum and mitochondria of yeast facilitates interorganelle transport of phospholipids through membrane contact. Eur J Biochem FEBS 264:545–553. doi:10.1046/j.1432-1327.1999.00658.x CrossRef Achleitner G, Gaigg B, Krasser A, Kainersdorfer E, Kohlwein SD, Perktold A, Zellnig G, Daum G (1999) Association between the endoplasmic reticulum and mitochondria of yeast facilitates interorganelle transport of phospholipids through membrane contact. Eur J Biochem FEBS 264:545–553. doi:10.​1046/​j.​1432-1327.​1999.​00658.​x CrossRef
5.
7.
Zurück zum Zitat Arruda AP, Pers BM, Parlakgül G, Güney E, Inouye K, Hotamisligil GS (2014) Chronic enrichment of hepatic endoplasmic reticulum-mitochondria contact leads to mitochondrial dysfunction in obesity. Nat Med 20:1427–1435. doi:10.1038/nm.3735 PubMedPubMedCentralCrossRef Arruda AP, Pers BM, Parlakgül G, Güney E, Inouye K, Hotamisligil GS (2014) Chronic enrichment of hepatic endoplasmic reticulum-mitochondria contact leads to mitochondrial dysfunction in obesity. Nat Med 20:1427–1435. doi:10.​1038/​nm.​3735 PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT (2008) Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol. doi:10.1083/jcb.200803137 PubMedPubMedCentral Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT (2008) Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol. doi:10.​1083/​jcb.​200803137 PubMedPubMedCentral
9.
Zurück zum Zitat Bernard-Marissal N, Medard JJ, Azzedine H, Chrast R (2015) Dysfunction in endoplasmic reticulum-mitochondria crosstalk underlies SIGMAR1 loss of function mediated motor neuron degeneration. Brain. doi:10.1093/brain/awv008 Bernard-Marissal N, Medard JJ, Azzedine H, Chrast R (2015) Dysfunction in endoplasmic reticulum-mitochondria crosstalk underlies SIGMAR1 loss of function mediated motor neuron degeneration. Brain. doi:10.​1093/​brain/​awv008
10.
Zurück zum Zitat Bernhard W, Rouiller C (1956) Close topographical relationship between mitochondria and ergastoplasm of liver cells in a definite phase of cellular activity. J Biophys Biochem Cytol 2:73–78PubMedPubMedCentralCrossRef Bernhard W, Rouiller C (1956) Close topographical relationship between mitochondria and ergastoplasm of liver cells in a definite phase of cellular activity. J Biophys Biochem Cytol 2:73–78PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Betz C, Stracka D, Prescianotto-Baschong C, Frieden M, Demaurex N, Hall MN (2013) mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology. Proc Natl Acad Sci 110:12526–12534. doi:10.1073/pnas.1302455110 PubMedPubMedCentralCrossRef Betz C, Stracka D, Prescianotto-Baschong C, Frieden M, Demaurex N, Hall MN (2013) mTOR complex 2-Akt signaling at mitochondria-associated endoplasmic reticulum membranes (MAM) regulates mitochondrial physiology. Proc Natl Acad Sci 110:12526–12534. doi:10.​1073/​pnas.​1302455110 PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Bouman L, Schlierf A, Lutz AK, Shan J, Deinlein A, Kast J, Galehdar Z, Palmisano V, Patenge N, Berg D et al (2011) Parkin is transcriptionally regulated by ATF4: evidence for an interconnection between mitochondrial stress and ER stress. Cell Death Differ 18:769–782. doi:10.1038/cdd.2010.142 PubMedPubMedCentralCrossRef Bouman L, Schlierf A, Lutz AK, Shan J, Deinlein A, Kast J, Galehdar Z, Palmisano V, Patenge N, Berg D et al (2011) Parkin is transcriptionally regulated by ATF4: evidence for an interconnection between mitochondrial stress and ER stress. Cell Death Differ 18:769–782. doi:10.​1038/​cdd.​2010.​142 PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Boyer O, Nevo F, Plaisier E, Funalot B, Gribouval O, Benoit G, Huynh Cong E, Arrondel C, Tete MJ, Montjean R et al (2011) INF2 mutations in Charcot–Marie–Tooth disease with glomerulopathy. N Engl J Med 365:2377–2388. doi:10.1056/NEJMoa1109122 PubMedCrossRef Boyer O, Nevo F, Plaisier E, Funalot B, Gribouval O, Benoit G, Huynh Cong E, Arrondel C, Tete MJ, Montjean R et al (2011) INF2 mutations in Charcot–Marie–Tooth disease with glomerulopathy. N Engl J Med 365:2377–2388. doi:10.​1056/​NEJMoa1109122 PubMedCrossRef
18.
Zurück zum Zitat Calì T, Ottolini D, Negro A, Brini M (2012) α-Synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J Biol Chem. doi:10.1074/jbc.M111.302794 Calì T, Ottolini D, Negro A, Brini M (2012) α-Synuclein controls mitochondrial calcium homeostasis by enhancing endoplasmic reticulum-mitochondria interactions. J Biol Chem. doi:10.​1074/​jbc.​M111.​302794
21.
Zurück zum Zitat Cheung K-HH, Mei L, Mak D-ODO, Hayashi I, Iwatsubo T, Kang DE, Foskett JK (2010) Gain-of-function enhancement of IP3 receptor modal gating by familial Alzheimer’s disease-linked presenilin mutants in human cells and mouse neurons. Sci Signal. doi:10.1126/scisignal.2000818 Cheung K-HH, Mei L, Mak D-ODO, Hayashi I, Iwatsubo T, Kang DE, Foskett JK (2010) Gain-of-function enhancement of IP3 receptor modal gating by familial Alzheimer’s disease-linked presenilin mutants in human cells and mouse neurons. Sci Signal. doi:10.​1126/​scisignal.​2000818
23.
Zurück zum Zitat Choo YS, Johnson GV, MacDonald M, Detloff PJ, Lesort M (2004) Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Hum Mol Genet 13:1407–1420. doi:10.1093/hmg/ddh162 PubMedCrossRef Choo YS, Johnson GV, MacDonald M, Detloff PJ, Lesort M (2004) Mutant huntingtin directly increases susceptibility of mitochondria to the calcium-induced permeability transition and cytochrome c release. Hum Mol Genet 13:1407–1420. doi:10.​1093/​hmg/​ddh162 PubMedCrossRef
24.
Zurück zum Zitat Copeland DE, Dalton AJ (1959) An association between mitochondria and the endoplasmic reticulum in cells of the pseudobranch gland of a teleost. J Biophys Biochem Cytol 5:393–396PubMedPubMedCentralCrossRef Copeland DE, Dalton AJ (1959) An association between mitochondria and the endoplasmic reticulum in cells of the pseudobranch gland of a teleost. J Biophys Biochem Cytol 5:393–396PubMedPubMedCentralCrossRef
25.
26.
Zurück zum Zitat Csordás G, Renken C, Várnai P, Walter L, Weaver D, Buttle KF, Balla T, Mannella CA, Hajnóczky G (2006) Structural and functional features and significance of the physical linkage between ER and mitochondria. J Cell Biol. doi:10.1083/jcb.200604016 PubMedPubMedCentral Csordás G, Renken C, Várnai P, Walter L, Weaver D, Buttle KF, Balla T, Mannella CA, Hajnóczky G (2006) Structural and functional features and significance of the physical linkage between ER and mitochondria. J Cell Biol. doi:10.​1083/​jcb.​200604016 PubMedPubMedCentral
29.
33.
Zurück zum Zitat Ferreira IL, Ferreiro E, Schmidt J, Cardoso JM, Pereira CMF, Carvalho AL, Oliveira CR, Rego ACR (2015) Aβ and NMDAR activation cause mitochondrial dysfunction involving ER calcium release. Neurobiol Aging 36:680692. doi:10.1016/j.neurobiolaging.2014.09.006 Ferreira IL, Ferreiro E, Schmidt J, Cardoso JM, Pereira CMF, Carvalho AL, Oliveira CR, Rego ACR (2015) Aβ and NMDAR activation cause mitochondrial dysfunction involving ER calcium release. Neurobiol Aging 36:680692. doi:10.​1016/​j.​neurobiolaging.​2014.​09.​006
34.
Zurück zum Zitat Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T, Pizzo P (2015) Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Natl Acad Sci USA 112:81. doi:10.1073/pnas.1504880112 CrossRef Filadi R, Greotti E, Turacchio G, Luini A, Pozzan T, Pizzo P (2015) Mitofusin 2 ablation increases endoplasmic reticulum-mitochondria coupling. Proc Natl Acad Sci USA 112:81. doi:10.​1073/​pnas.​1504880112 CrossRef
40.
Zurück zum Zitat Giacomello M, Drago I, Bortolozzi M, Scorzeto M, Gianelle A, Pizzo P, Pozzan T (2010) Ca2+ hot spots on the mitochondrial surface are generated by Ca2+ mobilization from stores, but not by activation of store-operated Ca2+ channels. Mol Cell. doi:10.1016/j.molcel.2010.04.003 PubMed Giacomello M, Drago I, Bortolozzi M, Scorzeto M, Gianelle A, Pizzo P, Pozzan T (2010) Ca2+ hot spots on the mitochondrial surface are generated by Ca2+ mobilization from stores, but not by activation of store-operated Ca2+ channels. Mol Cell. doi:10.​1016/​j.​molcel.​2010.​04.​003 PubMed
41.
Zurück zum Zitat Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A (2015) Glycogen synthase kinase-3 beta (GSK-3beta) signaling: implications for Parkinson’s disease. Pharmacol Res 97:16–26. doi:10.1016/j.phrs.2015.03.010 PubMedCrossRef Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A (2015) Glycogen synthase kinase-3 beta (GSK-3beta) signaling: implications for Parkinson’s disease. Pharmacol Res 97:16–26. doi:10.​1016/​j.​phrs.​2015.​03.​010 PubMedCrossRef
44.
Zurück zum Zitat Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y et al (2013) Autophagosomes form at ER-mitochondria contact sites. Nature 495:389–393. doi:10.1038/nature11910 PubMedCrossRef Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y et al (2013) Autophagosomes form at ER-mitochondria contact sites. Nature 495:389–393. doi:10.​1038/​nature11910 PubMedCrossRef
47.
Zurück zum Zitat Hedskog L, Pinho CM, Filadi R, Rönnbäck A, Hertwig L, Wiehager B, Larssen P, Gellhaar S, Sandebring A, Westerlund M et al (2013) Modulation of the endoplasmic reticulum–mitochondria interface in Alzheimer’s disease and related models. Proc Natl Acad Sci 110:7916–7921. doi:10.1073/pnas.1300677110 PubMedPubMedCentralCrossRef Hedskog L, Pinho CM, Filadi R, Rönnbäck A, Hertwig L, Wiehager B, Larssen P, Gellhaar S, Sandebring A, Westerlund M et al (2013) Modulation of the endoplasmic reticulum–mitochondria interface in Alzheimer’s disease and related models. Proc Natl Acad Sci 110:7916–7921. doi:10.​1073/​pnas.​1300677110 PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Joseph SK, Boehning D, Bokkala S, Watkins R, Widjaja J (1999) Biosynthesis of inositol trisphosphate receptors: selective association with the molecular chaperone calnexin. Biochem J 342(Pt 1):153–161PubMedPubMedCentralCrossRef Joseph SK, Boehning D, Bokkala S, Watkins R, Widjaja J (1999) Biosynthesis of inositol trisphosphate receptors: selective association with the molecular chaperone calnexin. Biochem J 342(Pt 1):153–161PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Kornak U, Mademan I, Schinke M, Voigt M, Krawitz P, Hecht J, Barvencik F, Schinke T, Gießelmann S, Beil TF et al (2014) Sensory neuropathy with bone destruction due to a mutation in the membrane-shaping atlastin GTPase 3. Brain 137:683–692. doi:10.1093/brain/awt357 PubMedCrossRef Kornak U, Mademan I, Schinke M, Voigt M, Krawitz P, Hecht J, Barvencik F, Schinke T, Gießelmann S, Beil TF et al (2014) Sensory neuropathy with bone destruction due to a mutation in the membrane-shaping atlastin GTPase 3. Brain 137:683–692. doi:10.​1093/​brain/​awt357 PubMedCrossRef
59.
Zurück zum Zitat Kremer A, Lippens S, Bartunkova S, Asselbergh B, Blanpain C, Fendrych M, Goossens A, Holt M, Janssens S, Krols M et al (2015) Developing 3D SEM in a broad biological context. J Microsci 259:80–96. doi:10.1111/jmi.12211 CrossRef Kremer A, Lippens S, Bartunkova S, Asselbergh B, Blanpain C, Fendrych M, Goossens A, Holt M, Janssens S, Krols M et al (2015) Developing 3D SEM in a broad biological context. J Microsci 259:80–96. doi:10.​1111/​jmi.​12211 CrossRef
60.
64.
Zurück zum Zitat Lim Y, Cho I-TT, Schoel LJ, Cho G, Golden JA (2015) Hereditary spastic paraplegia-linked reep1 modulates ER-mitochondria contacts. Ann Neurol. doi:10.1002/ana.24488 Lim Y, Cho I-TT, Schoel LJ, Cho G, Golden JA (2015) Hereditary spastic paraplegia-linked reep1 modulates ER-mitochondria contacts. Ann Neurol. doi:10.​1002/​ana.​24488
65.
Zurück zum Zitat Liu S, Sawada T, Lee S, Yu W, Silverio G, Alapatt P, Millan I, Shen A, Saxton W, Kanao T et al (2012) Parkinson’s disease-associated kinase PINK1 regulates Miro protein level and axonal transport of mitochondria. PLoS Genet. doi:10.1371/journal.pgen.1002537 Liu S, Sawada T, Lee S, Yu W, Silverio G, Alapatt P, Millan I, Shen A, Saxton W, Kanao T et al (2012) Parkinson’s disease-associated kinase PINK1 regulates Miro protein level and axonal transport of mitochondria. PLoS Genet. doi:10.​1371/​journal.​pgen.​1002537
67.
Zurück zum Zitat Luty AA, Kwok JBJ, Dobson-Stone C, Loy CT, Coupland KG, Karlström H, Sobow T, Tchorzewska J, Maruszak A, Barcikowska M et al (2010) Sigma nonopioid intracellular receptor 1 mutations cause frontotemporal lobar degeneration—motor neuron disease. Ann Neurol 68:639–649. doi:10.1002/ana.22274 PubMedCrossRef Luty AA, Kwok JBJ, Dobson-Stone C, Loy CT, Coupland KG, Karlström H, Sobow T, Tchorzewska J, Maruszak A, Barcikowska M et al (2010) Sigma nonopioid intracellular receptor 1 mutations cause frontotemporal lobar degeneration—motor neuron disease. Ann Neurol 68:639–649. doi:10.​1002/​ana.​22274 PubMedCrossRef
70.
Zurück zum Zitat Manor U, Bartholomew S, Golani G, Christenson E, Kozlov M, Higgs H, Spudich J, Lippincott-Schwartz J (2015) A mitochondria-anchored isoform of the actin-nucleating Spire protein regulates mitochondrial division. eLife 4. doi:10.7554/eLife.08828 Manor U, Bartholomew S, Golani G, Christenson E, Kozlov M, Higgs H, Spudich J, Lippincott-Schwartz J (2015) A mitochondria-anchored isoform of the actin-nucleating Spire protein regulates mitochondrial division. eLife 4. doi:10.​7554/​eLife.​08828
77.
Zurück zum Zitat Mórotz GM, De Vos KJ, Vagnoni A, Ackerley S, Shaw CE, Miller CC (2012) Amyotrophic lateral sclerosis-associated mutant VAPBP56S perturbs calcium homeostasis to disrupt axonal transport of mitochondria. Hum Mol Genet 21:1979–1988. doi:10.1093/hmg/dds011 PubMedPubMedCentralCrossRef Mórotz GM, De Vos KJ, Vagnoni A, Ackerley S, Shaw CE, Miller CC (2012) Amyotrophic lateral sclerosis-associated mutant VAPBP56S perturbs calcium homeostasis to disrupt axonal transport of mitochondria. Hum Mol Genet 21:1979–1988. doi:10.​1093/​hmg/​dds011 PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Nguyen TT, Oh SS, Weaver D, Lewandowska A, Maxfield D, Schuler M-HH, Smith NK, Macfarlane J, Saunders G, Palmer CA et al (2014) Loss of Miro1-directed mitochondrial movement results in a novel murine model for neuron disease. Proc Natl Acad Sci USA 111:40. doi:10.1073/pnas.1402449111 CrossRef Nguyen TT, Oh SS, Weaver D, Lewandowska A, Maxfield D, Schuler M-HH, Smith NK, Macfarlane J, Saunders G, Palmer CA et al (2014) Loss of Miro1-directed mitochondrial movement results in a novel murine model for neuron disease. Proc Natl Acad Sci USA 111:40. doi:10.​1073/​pnas.​1402449111 CrossRef
81.
Zurück zum Zitat Nishimura AL, Mitne-Neto M, Silva HC, Richieri-Costa A, Middleton S, Cascio D, Kok F, Oliveira JR, Gillingwater T, Webb J et al (2004) A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75:822–831. doi:10.1086/425287 PubMedPubMedCentralCrossRef Nishimura AL, Mitne-Neto M, Silva HC, Richieri-Costa A, Middleton S, Cascio D, Kok F, Oliveira JR, Gillingwater T, Webb J et al (2004) A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis. Am J Hum Genet 75:822–831. doi:10.​1086/​425287 PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Ottolini D, Calì T, Negro A, Brini M (2013) The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum–mitochondria tethering. Hum Mol Genet 22:2152–2168. doi:10.1093/hmg/ddt068 PubMedCrossRef Ottolini D, Calì T, Negro A, Brini M (2013) The Parkinson disease-related protein DJ-1 counteracts mitochondrial impairment induced by the tumour suppressor protein p53 by enhancing endoplasmic reticulum–mitochondria tethering. Hum Mol Genet 22:2152–2168. doi:10.​1093/​hmg/​ddt068 PubMedCrossRef
83.
Zurück zum Zitat Panov AV, Gutekunst C-AA, Leavitt BR, Hayden MR, Burke JR, Strittmatter WJ, Greenamyre JT (2002) Early mitochondrial calcium defects in Huntington’s disease are a direct effect of polyglutamines. Nat Neurosci 5:731–736. doi:10.1038/nn884 PubMed Panov AV, Gutekunst C-AA, Leavitt BR, Hayden MR, Burke JR, Strittmatter WJ, Greenamyre JT (2002) Early mitochondrial calcium defects in Huntington’s disease are a direct effect of polyglutamines. Nat Neurosci 5:731–736. doi:10.​1038/​nn884 PubMed
84.
Zurück zum Zitat Pivovarova NB, Pozzo-Miller LD, Hongpaisan J, Andrews SB (2002) Correlated calcium uptake and release by mitochondria and endoplasmic reticulum of CA3 hippocampal dendrites after afferent synaptic stimulation. J Neurosci 22:10653–10661PubMed Pivovarova NB, Pozzo-Miller LD, Hongpaisan J, Andrews SB (2002) Correlated calcium uptake and release by mitochondria and endoplasmic reticulum of CA3 hippocampal dendrites after afferent synaptic stimulation. J Neurosci 22:10653–10661PubMed
85.
Zurück zum Zitat Prause J, Goswami A, Katona I, Roos A, Schnizler M, Bushuven E, Dreier A, Buchkremer S, Johann S, Beyer C et al (2013) Altered localization, abnormal modification and loss of function of Sigma receptor-1 in amyotrophic lateral sclerosis. Hum Mol Genet 22:1581–1600. doi:10.1093/hmg/ddt008 PubMedCrossRef Prause J, Goswami A, Katona I, Roos A, Schnizler M, Bushuven E, Dreier A, Buchkremer S, Johann S, Beyer C et al (2013) Altered localization, abnormal modification and loss of function of Sigma receptor-1 in amyotrophic lateral sclerosis. Hum Mol Genet 22:1581–1600. doi:10.​1093/​hmg/​ddt008 PubMedCrossRef
87.
89.
97.
Zurück zum Zitat Stoica R, De Vos KJ, Paillusson S, Mueller S, Sancho RM, Lau K-FF, Vizcay-Barrena G, Lin W-LL, Xu Y-FF, Lewis J et al (2014) ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat Commun 5:3996. doi:10.1038/ncomms4996 PubMedPubMedCentral Stoica R, De Vos KJ, Paillusson S, Mueller S, Sancho RM, Lau K-FF, Vizcay-Barrena G, Lin W-LL, Xu Y-FF, Lewis J et al (2014) ER-mitochondria associations are regulated by the VAPB-PTPIP51 interaction and are disrupted by ALS/FTD-associated TDP-43. Nat Commun 5:3996. doi:10.​1038/​ncomms4996 PubMedPubMedCentral
99.
100.
Zurück zum Zitat Szabadkai G, Bianchi K, Várnai P, De Stefani D, Wieckowski MR, Cavagna D, Nagy AI, Balla T, Rizzuto R (2006) Chaperone-mediated coupling of endoplasmic reticulum and mitochondrial Ca2+ channels. J Cell Biol. doi:10.1083/jcb.200608073 Szabadkai G, Bianchi K, Várnai P, De Stefani D, Wieckowski MR, Cavagna D, Nagy AI, Balla T, Rizzuto R (2006) Chaperone-mediated coupling of endoplasmic reticulum and mitochondrial Ca2+ channels. J Cell Biol. doi:10.​1083/​jcb.​200608073
104.
Zurück zum Zitat Tubbs E, Theurey P, Vial G, Bendridi N, Bravard A, Chauvin M-A, Ji-Cao J, Zoulim F, Bartosch B, Ovize M et al (2014) Mitochondria-associated endoplasmic reticulum membrane (MAM) integrity is required for insulin signaling and is implicated in hepatic insulin resistance. Diabetes 63:3279–3294. doi:10.2337/db13-1751 PubMedCrossRef Tubbs E, Theurey P, Vial G, Bendridi N, Bravard A, Chauvin M-A, Ji-Cao J, Zoulim F, Bartosch B, Ovize M et al (2014) Mitochondria-associated endoplasmic reticulum membrane (MAM) integrity is required for insulin signaling and is implicated in hepatic insulin resistance. Diabetes 63:3279–3294. doi:10.​2337/​db13-1751 PubMedCrossRef
106.
107.
Zurück zum Zitat Van Laar VS, Roy N, Liu A, Rajprohat S, Arnold B, Dukes AA, Holbein CD, Berman SB (2014) Glutamate excitotoxicity in neurons triggers mitochondrial and endoplasmic reticulum accumulation of Parkin, and in the presence of N-acetyl cysteine, mitophagy. Neurobiol Dis. doi:10.1016/j.nbd.2014.11.015 PubMedPubMedCentral Van Laar VS, Roy N, Liu A, Rajprohat S, Arnold B, Dukes AA, Holbein CD, Berman SB (2014) Glutamate excitotoxicity in neurons triggers mitochondrial and endoplasmic reticulum accumulation of Parkin, and in the presence of N-acetyl cysteine, mitophagy. Neurobiol Dis. doi:10.​1016/​j.​nbd.​2014.​11.​015 PubMedPubMedCentral
110.
Zurück zum Zitat Vance JE (1990) Phospholipid synthesis in a membrane fraction associated with mitochondria. J Biol Chem 265:7248–7256PubMed Vance JE (1990) Phospholipid synthesis in a membrane fraction associated with mitochondria. J Biol Chem 265:7248–7256PubMed
111.
Zurück zum Zitat Vishnu N, Jadoon Khan M, Karsten F, Groschner LN, Waldeck-Weiermair M, Rost R, Hallström S, Imamura H, Graier WF, Malli R (2014) ATP increases within the lumen of the endoplasmic reticulum upon intracellular Ca2+ release. Mol Biol Cell 25:368–379. doi:10.1091/mbc.E13-07-0433 PubMedPubMedCentralCrossRef Vishnu N, Jadoon Khan M, Karsten F, Groschner LN, Waldeck-Weiermair M, Rost R, Hallström S, Imamura H, Graier WF, Malli R (2014) ATP increases within the lumen of the endoplasmic reticulum upon intracellular Ca2+ release. Mol Biol Cell 25:368–379. doi:10.​1091/​mbc.​E13-07-0433 PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Voelker DR (1989) Phosphatidylserine translocation to the mitochondrion is an ATP-dependent process in permeabilized animal cells. Proc Natl Acad Sci USA 86:9921–9925PubMedPubMedCentralCrossRef Voelker DR (1989) Phosphatidylserine translocation to the mitochondrion is an ATP-dependent process in permeabilized animal cells. Proc Natl Acad Sci USA 86:9921–9925PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Wiley SE, Andreyev AY, Divakaruni AS, Karisch R, Perkins G, Wall EA, van der Geer P, Chen Y-FF, Tsai T-FF, Simon MI et al (2013) Wolfram Syndrome protein, Miner1, regulates sulphydryl redox status, the unfolded protein response, and Ca2+ homeostasis. EMBO Mol Med 5:904–918. doi:10.1002/emmm.201201429 PubMedPubMedCentralCrossRef Wiley SE, Andreyev AY, Divakaruni AS, Karisch R, Perkins G, Wall EA, van der Geer P, Chen Y-FF, Tsai T-FF, Simon MI et al (2013) Wolfram Syndrome protein, Miner1, regulates sulphydryl redox status, the unfolded protein response, and Ca2+ homeostasis. EMBO Mol Med 5:904–918. doi:10.​1002/​emmm.​201201429 PubMedPubMedCentralCrossRef
Metadaten
Titel
Mitochondria-associated membranes as hubs for neurodegeneration
verfasst von
Michiel Krols
Gert van Isterdael
Bob Asselbergh
Anna Kremer
Saskia Lippens
Vincent Timmerman
Sophie Janssens
Publikationsdatum
01.04.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Acta Neuropathologica / Ausgabe 4/2016
Print ISSN: 0001-6322
Elektronische ISSN: 1432-0533
DOI
https://doi.org/10.1007/s00401-015-1528-7

Weitere Artikel der Ausgabe 4/2016

Acta Neuropathologica 4/2016 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Update Neurologie

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