Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 552
Filter
1.
Immunity ; 52(5): 767-781.e6, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32277911

ABSTRACT

The enzyme cyclic GMP-AMP synthase (cGAS) senses cytosolic DNA in infected and malignant cells and catalyzes the formation of 2'3'cGMP-AMP (cGAMP), which in turn triggers interferon (IFN) production via the STING pathway. Here, we examined the contribution of anion channels to cGAMP transfer and anti-viral defense. A candidate screen revealed that inhibition of volume-regulated anion channels (VRACs) increased propagation of the DNA virus HSV-1 but not the RNA virus VSV. Chemical blockade or genetic ablation of LRRC8A/SWELL1, a VRAC subunit, resulted in defective IFN responses to HSV-1. Biochemical and electrophysiological analyses revealed that LRRC8A/LRRC8E-containing VRACs transport cGAMP and cyclic dinucleotides across the plasma membrane. Enhancing VRAC activity by hypotonic cell swelling, cisplatin, GTPγS, or the cytokines TNF or interleukin-1 increased STING-dependent IFN response to extracellular but not intracellular cGAMP. Lrrc8e-/- mice exhibited impaired IFN responses and compromised immunity to HSV-1. Our findings suggest that cell-to-cell transmission of cGAMP via LRRC8/VRAC channels is central to effective anti-viral immunity.


Subject(s)
Fibroblasts/immunology , Interferons/immunology , Membrane Proteins/immunology , Nucleotides, Cyclic/immunology , Voltage-Dependent Anion Channels/immunology , Animals , Antiviral Agents/immunology , Antiviral Agents/metabolism , Bystander Effect , Cell Line , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Humans , Interferons/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Voltage-Dependent Anion Channels/metabolism
2.
Dev Biol ; 513: 50-62, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38492873

ABSTRACT

The voltage gated (Kv) slow-inactivating delayed rectifier channel regulates the development of hollow organs of the zebrafish. The functional channel consists of the tetramer of electrically active Kcnb1 (Kv2.1) subunits and Kcng4b (Kv6.4) modulatory or electrically silent subunits. The two mutations in zebrafish kcng4b gene - kcng4b-C1 and kcng4b-C2 (Gasanov et al., 2021) - have been studied during ear development using electrophysiology, developmental biology and in silico structural modelling. kcng4b-C1 mutation causes a C-terminal truncation characterized by mild Kcng4b loss-of-function (LOF) manifested by failure of kinocilia to extend and formation of ectopic otoliths. In contrast, the kcng4b-C2-/- mutation causes the C-terminal domain to elongate and the ectopic seventh transmembrane (TM) domain to form, converting the intracellular C-terminus to an extracellular one. Kcng4b-C2 acts as a Kcng4b gain-of-function (GOF) allele. Otoliths fail to develop and kinocilia are reduced in kcng4b-C2-/-. These results show that different mutations of the silent subunit Kcng4 can affect the activity of the Kv channel and cause a wide range of developmental defects.


Subject(s)
Ear , Voltage-Dependent Anion Channels , Zebrafish Proteins , Zebrafish , Animals , Ear/embryology , Mutation/genetics , Zebrafish/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Voltage-Dependent Anion Channels/genetics , Voltage-Dependent Anion Channels/metabolism
3.
Plant Physiol ; 194(2): 1041-1058, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-37772952

ABSTRACT

In Arabidopsis (Arabidopsis thaliana), stomatal closure mediated by abscisic acid (ABA) is redundantly controlled by ABA receptor family proteins (PYRABACTIN RESISTANCE 1 [PYR1]/PYR1-LIKE [PYLs]) and subclass III SUCROSE NONFERMENTING 1 (SNF1)-RELATED PROTEIN KINASES 2 (SnRK2s). Among these proteins, the roles of PYR1, PYL2, and SnRK2.6 are more dominant. A recent discovery showed that ABA-induced accumulation of reactive oxygen species (ROS) in mitochondria promotes stomatal closure. By analyzing stomatal movements in an array of single and higher order mutants, we revealed that the mitochondrial protein VOLTAGE-DEPENDENT ANION CHANNEL 3 (VDAC3) jointly regulates ABA-mediated stomatal closure with a specialized set of PYLs and SnRK2s by affecting cellular and mitochondrial ROS accumulation. VDAC3 interacted with 9 PYLs and all 3 subclass III SnRK2s. Single mutation in VDAC3, PYLs (except PYR1 and PYL2), or SnRK2.2/2.3 had little effect on ABA-mediated stomatal closure. However, knocking out PYR1, PYL1/2/4/8, or SnRK2.2/2.3 in vdac3 mutants resulted in significantly delayed or attenuated ABA-mediated stomatal closure, despite the presence of other PYLs or SnRK2s conferring redundant functions. We found that cellular and mitochondrial accumulation of ROS induced by ABA was altered in vdac3pyl1 mutants. Moreover, H2O2 treatment restored ABA-induced stomatal closure in mutants with decreased stomatal sensitivity to ABA. Our work reveals that VDAC3 ensures redundant control of ABA-mediated stomatal closure by canonical ABA signaling components.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Abscisic Acid/pharmacology , Abscisic Acid/metabolism , Arabidopsis Proteins/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Plant Stomata/metabolism , Arabidopsis/metabolism , Voltage-Dependent Anion Channels/metabolism , Mitochondria/metabolism
4.
Arch Biochem Biophys ; 753: 109914, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38290597

ABSTRACT

The mitochondrial outer membrane protein porin 1 (Por1), the yeast orthologue of mammalian voltage-dependent anion channel (VDAC), is the major permeability pathway for the flux of metabolites and ions between cytosol and mitochondria. In yeast, several Por1 phosphorylation sites have been identified. Protein phosphorylation is a major modification regulating a variety of biological activities, but the potential biological roles of Por1 phosphorylation remains unaddressed. In this work, we analysed 10 experimentally observed phosphorylation sites in yeast Por1 using bioinformatics tools. Two of the residues, T100 and S133, predicted to reduce and increase pore permeability, respectively, were validated using biological assays. In accordance, Por1T100D reduced mitochondrial respiration, while Por1S133E phosphomimetic mutant increased it. Por1T100A expression also improved respiratory growth, while Por1S133A caused defects in all growth conditions tested, notably in fermenting media. In conclusion, we found phosphorylation has the potential to modulate Por1, causing a marked effect on mitochondrial function. It can also impact on cell morphology and growth both in respiratory and, unpredictably, also in fermenting conditions, expanding our knowledge on the role of Por1 in cell physiology.


Subject(s)
Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Animals , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Phosphorylation , Voltage-Dependent Anion Channels/metabolism , Mitochondria/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Mammals/metabolism
5.
Rev Med Virol ; 33(4): e2453, 2023 07.
Article in English | MEDLINE | ID: mdl-37170417

ABSTRACT

Viruses control the host cell by exploiting its molecular machinery to facilitate viral replication and propagation. Understanding different viral mechanisms and biochemical pathways is crucial for finding promising therapeutic solutions to viral infections. The mitochondrion is a vital organelle targeted by various types of viruses. More specifically, viruses interact with the voltage-dependent anion channel (VDAC), a porin protein found in the outer mitochondrial membrane. VDAC controls metabolite flux, regulates reactive oxygen species production, and promotes mitochondrial-mediated apoptosis by releasing pro-apoptotic proteins. Hence, a common pathogenic strategy used by many viruses seems to exploit natural pathways that VDAC regulates. This review aims to address the inhibition and enhancement roles of VDAC in viral pathogenesis and outlines multiple links and interactions between VDAC and viral proteins as potential antiviral targets.


Subject(s)
Virus Diseases , Voltage-Dependent Anion Channels , Humans , Voltage-Dependent Anion Channels/metabolism , Mitochondria/metabolism , Apoptosis , Viral Proteins/metabolism , Virus Diseases/metabolism
6.
Biochemistry (Mosc) ; 89(6): 1061-1078, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38981701

ABSTRACT

Voltage-dependent anion channels (VDAC1-3) of the outer mitochondrial membrane are a family of pore-forming ß-barrel proteins that carry out controlled "filtration" of small molecules and ions between the cytoplasm and mitochondria. Due to the conformational transitions between the closed and open states and interaction with cytoplasmic and mitochondrial proteins, VDACs not only regulate the mitochondrial membrane permeability for major metabolites and ions, but also participate in the control of essential intracellular processes and pathological conditions. This review discusses novel data on the molecular structure, regulatory mechanisms, and pathophysiological role of VDAC proteins, as well as future directions in this area of research.


Subject(s)
Mitochondrial Membranes , Voltage-Dependent Anion Channels , Humans , Voltage-Dependent Anion Channels/metabolism , Mitochondrial Membranes/metabolism , Animals , Mitochondria/metabolism
7.
Int J Mol Sci ; 25(5)2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38474278

ABSTRACT

The small GTPase RAS acts as a plasma membrane-anchored intracellular neurotrophin counteracting neuronal degeneration in the brain, but the underlying molecular mechanisms are largely unknown. In transgenic mice expressing constitutively activated V12-Ha-RAS selectively in neurons, proteome analysis uncovered a 70% decrease in voltage-dependent anion channel-1 (VDAC-1) in the cortex and hippocampus. We observed a corresponding reduction in the levels of mRNA splicing variant coding for plasma membrane-targeted VDAC-1 (pl-VDAC-1) while mRNA levels for mitochondrial membrane VDAC-1 (mt-VDAC-1) remained constant. In primary cortical neurons derived from V12-Ha-RAS animals, a decrease in pl-VDAC-1 mRNA levels was observed, accompanied by a concomitant reduction in the ferricyanide reductase activity associated with VDAC-1 protein. Application of MEK inhibitor U0126 to transgenic cortical neurons reconstituted pl-VDAC-1 mRNA to reach wild-type levels. Excitotoxic glutamate-induced cell death was strongly attenuated in transgenic V12-Ha-RAS overexpressing cortical cultures. Consistently, a neuroprotective effect could also be achieved in wild-type cortical cultures by the extracellular application of channel-blocking antibody targeting the N-terminus of VDAC-1. These results may encourage novel therapeutic approaches toward blocking pl-VDAC-1 by monoclonal antibody targeting for complementary treatments in transplantation and neurodegenerative disease.


Subject(s)
Neurodegenerative Diseases , Voltage-Dependent Anion Channels , Mice , Animals , Voltage-Dependent Anion Channels/metabolism , Neuroprotection , Neurodegenerative Diseases/metabolism , ras Proteins/metabolism , Down-Regulation , Voltage-Dependent Anion Channel 1/metabolism , Cell Membrane/metabolism , Mice, Transgenic , RNA, Messenger/metabolism , Voltage-Dependent Anion Channel 2/metabolism
8.
Plant J ; 109(1): 241-260, 2022 01.
Article in English | MEDLINE | ID: mdl-34748255

ABSTRACT

Calcium (Ca2+ ) is widely recognized as a key second messenger in mediating various plant adaptive responses. Here we show that calcineurin B-like interacting protein kinase CIPK9 along with its interacting partner VDAC3 identified in the present study are involved in mediating plant responses to methyl viologen (MV). CIPK9 physically interacts with and phosphorylates VDAC3. Co-localization, co-immunoprecipitation, and fluorescence resonance energy transfer experiments proved their physical interaction in planta. Both cipk9 and vdac3 mutants exhibited a tolerant phenotype against MV-induced oxidative stress, which coincided with the lower-level accumulation of reactive oxygen species in their roots. In addition, the analysis of cipk9vdac3 double mutant and VDAC3 overexpressing plants revealed that CIPK9 and VDAC3 were involved in the same pathway for inducing MV-dependent oxidative stress. The response to MV was suppressed by the addition of lanthanum chloride, a non-specific Ca2+ channel blocker indicating the role of Ca2+ in this pathway. Our study suggest that CIPK9-VDAC3 module may act as a key component in mediating oxidative stress responses in Arabidopsis.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Protein Serine-Threonine Kinases/metabolism , Voltage-Dependent Anion Channels/metabolism , Arabidopsis/drug effects , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Gene Expression Regulation, Plant , Lanthanum/pharmacology , Oxidative Stress , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Voltage-Dependent Anion Channels/genetics
9.
J Cell Sci ; 134(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34523684

ABSTRACT

The voltage-dependent anion channel (VDAC) is a ubiquitous channel in the outer membrane of the mitochondrion with multiple roles in protein, metabolite and small molecule transport. In mammalian cells, VDAC protein, as part of a larger complex including the inositol triphosphate receptor, has been shown to have a role in mediating contacts between the mitochondria and endoplasmic reticulum (ER). We identify VDAC of the pathogenic apicomplexan Toxoplasma gondii and demonstrate its importance for parasite growth. We show that VDAC is involved in protein import and metabolite transfer to mitochondria. Further, depletion of VDAC resulted in significant morphological changes in the mitochondrion and ER, suggesting a role in mediating contacts between these organelles in T. gondii. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Toxoplasma , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Mitochondria/metabolism , Protein Transport , Toxoplasma/genetics , Toxoplasma/metabolism , Voltage-Dependent Anion Channels/genetics , Voltage-Dependent Anion Channels/metabolism
10.
Chembiochem ; 24(14): e202300111, 2023 07 17.
Article in English | MEDLINE | ID: mdl-36964942

ABSTRACT

Chlorinated gymnastatin and dankastatin alkaloids derived from the fungal strain Gymnascella dankaliensis have been reported to possess significant anticancer activity but their mode of action is unknown. These members possess electrophilic functional groups that can might undergo covalent bond formation with specific proteins to exert their biological activity. To better understand the mechanism of action of this class of natural products, we mapped the proteome-wide cysteine reactivity of the most potent of these alkaloids, dankastatin B, by using activity-based protein profiling chemoproteomic approaches. We identified a primary target of dankastatin B in breast cancer cells as cysteine C65 of the voltage-dependent anion-selective channel on the outer mitochondrial membrane VDAC3. We demonstrated direct and covalent interaction of dankastatin B with VDAC3. VDAC3 knockdown conferred hypersensitivity to dankastatin B-mediated antiproliferative effects in breast cancer cells, thus indicating that VDAC3 was at least partially involved in the anticancer effects of this natural product. Our study reveals a potential mode of action of dankastatin B through covalent targeting of VDAC3 and highlights the utility of chemoproteomic approaches in gaining mechanistic understanding of electrophilic natural products.


Subject(s)
Biological Products , Breast Neoplasms , Humans , Female , Cysteine/chemistry , Biological Products/chemistry , Mitochondria/metabolism , Breast Neoplasms/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Voltage-Dependent Anion Channels/metabolism
11.
Plant Physiol ; 188(2): 1061-1080, 2022 02 04.
Article in English | MEDLINE | ID: mdl-34747475

ABSTRACT

Infection cycles of viruses are highly dependent on membrane-associated host factors. To uncover the infection cycle of Bamboo mosaic virus (BaMV) in detail, we purified the membrane-associated viral complexes from infected Nicotiana benthamiana plants and analyzed the involved host factors. Four isoforms of voltage-dependent anion channel (VDAC) proteins on the outer membrane of mitochondria were identified due to their upregulated expression in the BaMV complex-enriched membranous fraction. Results from loss- and gain-of-function experiments indicated that NbVDAC2, -3, and -4 are essential for efficient BaMV accumulation. During BaMV infection, all NbVDACs concentrated into larger aggregates, which overlapped and trafficked with BaMV virions to the structure designated as the "dynamic BaMV-induced complex." Besides the endoplasmic reticulum and mitochondria, BaMV replicase and double-stranded RNAs were also found in this complex, suggesting the dynamic BaMV-induced complex is a replication complex. Yeast two-hybrid and pull-down assays confirmed that BaMV triple gene block protein 1 (TGBp1) could interact with NbVDACs. Confocal microscopy revealed that TGBp1 is sufficient to induce NbVDAC aggregates, which suggests that TGBp1 may play a pivotal role in the NbVDAC-virion complex. Collectively, these findings indicate that NbVDACs may associate with the dynamic BaMV-induced complex via TGBp1 and NbVDAC2, -3, or -4 and can promote BaMV accumulation. This study reveals the involvement of mitochondrial proteins in a viral complex and virus infection.


Subject(s)
Membrane Proteins/metabolism , Mosaic Viruses/pathogenicity , Nicotiana/virology , Plant Diseases/virology , Potexvirus/pathogenicity , RNA-Dependent RNA Polymerase/metabolism , Voltage-Dependent Anion Channels/metabolism , Host-Parasite Interactions
12.
Cell Mol Life Sci ; 79(7): 368, 2022 Jun 19.
Article in English | MEDLINE | ID: mdl-35718804

ABSTRACT

Involvement of alpha-synuclein (αSyn) in Parkinson's disease (PD) is complicated and difficult to trace on cellular and molecular levels. Recently, we established that αSyn can regulate mitochondrial function by voltage-activated complexation with the voltage-dependent anion channel (VDAC) on the mitochondrial outer membrane. When complexed with αSyn, the VDAC pore is partially blocked, reducing the transport of ATP/ADP and other metabolites. Further, αSyn can translocate into the mitochondria through VDAC, where it interferes with mitochondrial respiration. Recruitment of αSyn to the VDAC-containing lipid membrane appears to be a crucial prerequisite for both the blockage and translocation processes. Here we report an inhibitory effect of HK2p, a small membrane-binding peptide from the mitochondria-targeting N-terminus of hexokinase 2, on αSyn membrane binding, and hence on αSyn complex formation with VDAC and translocation through it. In electrophysiology experiments, the addition of HK2p at micromolar concentrations to the same side of the membrane as αSyn results in a dramatic reduction of the frequency of blockage events in a concentration-dependent manner, reporting on complexation inhibition. Using two complementary methods of measuring protein-membrane binding, bilayer overtone analysis and fluorescence correlation spectroscopy, we found that HK2p induces detachment of αSyn from lipid membranes. Experiments with HeLa cells using proximity ligation assay confirmed that HK2p impedes αSyn entry into mitochondria. Our results demonstrate that it is possible to regulate αSyn-VDAC complexation by a rationally designed peptide, thus suggesting new avenues in the search for peptide therapeutics to alleviate αSyn mitochondrial toxicity in PD and other synucleinopathies.


Subject(s)
Parkinson Disease , alpha-Synuclein , HeLa Cells , Humans , Lipids , Mitochondria/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Voltage-Dependent Anion Channels/metabolism , alpha-Synuclein/metabolism
13.
Biol Res ; 56(1): 33, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37344914

ABSTRACT

BACKGROUND: Voltage-dependent anion selective channels (VDACs) are the most abundant mitochondrial outer membrane proteins, encoded in mammals by three genes, VDAC1, 2 and 3, mostly ubiquitously expressed. As 'mitochondrial gatekeepers', VDACs control organelle and cell metabolism and are involved in many diseases. Despite the presence of numerous VDAC pseudogenes in the human genome, their significance and possible role in VDAC protein expression has not yet been considered. RESULTS: We investigated the relevance of processed pseudogenes of human VDAC genes, both in physiological and in pathological contexts. Using high-throughput tools and querying many genomic and transcriptomic databases, we show that some VDAC pseudogenes are transcribed in specific tissues and pathological contexts. The obtained experimental data confirm an association of the VDAC1P8 pseudogene with acute myeloid leukemia (AML). CONCLUSIONS: Our in-silico comparative analysis between the VDAC1 gene and its VDAC1P8 pseudogene, together with experimental data produced in AML cellular models, indicate a specific over-expression of the VDAC1P8 pseudogene in AML, correlated with a downregulation of the parental VDAC1 gene.


Subject(s)
Leukemia, Myeloid, Acute , Pseudogenes , Voltage-Dependent Anion Channels , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mitochondria , Pseudogenes/genetics , Transcriptome , Voltage-Dependent Anion Channels/genetics , Voltage-Dependent Anion Channels/metabolism
14.
Proc Natl Acad Sci U S A ; 117(35): 21740-21746, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817533

ABSTRACT

The Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) anion channel is essential for epithelial salt-water balance. CFTR mutations cause cystic fibrosis, a lethal incurable disease. In cells CFTR is activated through the cAMP signaling pathway, overstimulation of which during cholera leads to CFTR-mediated intestinal salt-water loss. Channel activation is achieved by phosphorylation of its regulatory (R) domain by cAMP-dependent protein kinase catalytic subunit (PKA). Here we show using two independent approaches--an ATP analog that can drive CFTR channel gating but is unsuitable for phosphotransfer by PKA, and CFTR mutants lacking phosphorylatable serines--that PKA efficiently opens CFTR channels through simple binding, under conditions that preclude phosphorylation. Unlike when phosphorylation happens, CFTR activation by PKA binding is completely reversible. Thus, PKA binding promotes release of the unphosphorylated R domain from its inhibitory position, causing full channel activation, whereas phosphorylation serves only to maintain channel activity beyond termination of the PKA signal. The results suggest two levels of CFTR regulation in cells: irreversible through phosphorylation, and reversible through R-domain binding to PKA--and possibly also to other members of a large network of proteins known to interact with the channel.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Voltage-Dependent Anion Channels/metabolism , Adenosine Triphosphate/metabolism , Animals , Anions/metabolism , Biophysical Phenomena , Cyclic AMP-Dependent Protein Kinases/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Ion Channel Gating/physiology , Mutagenesis, Site-Directed , Nucleotides/metabolism , Oocytes/metabolism , Patch-Clamp Techniques/methods , Phosphorylation , Protein Binding/physiology , Serine/metabolism , Voltage-Dependent Anion Channels/physiology , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
15.
Int J Mol Sci ; 24(7)2023 Apr 02.
Article in English | MEDLINE | ID: mdl-37047622

ABSTRACT

The voltage-dependent anion channel (VDAC) is the primary regulating pathway of water-soluble metabolites and ions across the mitochondrial outer membrane [...].


Subject(s)
Mitochondrial Membranes , Voltage-Dependent Anion Channels , Voltage-Dependent Anion Channels/metabolism , Mitochondrial Membranes/metabolism , Mitochondria/metabolism , Ions/metabolism
16.
Molecules ; 28(8)2023 Apr 08.
Article in English | MEDLINE | ID: mdl-37110543

ABSTRACT

VDAC (Voltage-Dependent Anion-selective Channel) is the primary metabolite pore in the mitochondrial outer membrane (OM). Atomic structures of VDAC, consistent with its physiological "open" state, are ß-barrels formed by 19 transmembrane (TM) ß-strands and an N-terminal segment (NTERM) that folds inside the pore lumen. However, structures are lacking for VDAC's partially "closed" states. To provide clues about possible VDAC conformers, we used the RoseTTAFold neural network to predict structures for human and fungal VDAC sequences modified to mimic removal from the pore wall or lumen of "cryptic" domains, i.e., segments buried in atomic models yet accessible to antibodies in OM-bound VDAC. Predicted in vacuo structures for full-length VDAC sequences are 19-strand ß-barrels similar to atomic models, but with weaker H-bonding between TM strands and reduced interactions between NTERM and the pore wall. Excision of combinations of "cryptic" subregions yields ß-barrels with smaller diameters, wide gaps between N- and C-terminal ß-strands, and in some cases disruption of the ß-sheet (associated with strained backbone H-bond registration). Tandem repeats of modified VDAC sequences also were explored, as was domain swapping in monomer constructs. Implications of the results for possible alternative conformational states of VDAC are discussed.


Subject(s)
Mitochondrial Membranes , Voltage-Dependent Anion Channels , Humans , Mitochondrial Membranes/metabolism , Voltage-Dependent Anion Channels/metabolism , Molecular Conformation
17.
Proteomics ; 22(5-6): e2100060, 2022 03.
Article in English | MEDLINE | ID: mdl-34813679

ABSTRACT

Voltage-activated complexation is the process by which a transmembrane potential drives complex formation between a membrane-embedded channel and a soluble or membrane-peripheral target protein. Metabolite and calcium flux across the mitochondrial outer membrane was shown to be regulated by voltage-activated complexation of the voltage-dependent anion channel (VDAC) and either dimeric tubulin or α-synuclein (αSyn). However, the roles played by VDAC's characteristic attributes-its anion selectivity and voltage gating behavior-have remained unclear. Here, we compare in vitro measurements of voltage-activated complexation of αSyn with three well-characterized ß-barrel channels-VDAC, MspA, and α-hemolysin-that differ widely in their organism of origin, structure, geometry, charge density distribution, and voltage gating behavior. The voltage dependences of the complexation dynamics for the different channels are observed to differ quantitatively but have similar qualitative features. In each case, energy landscape modeling describes the complexation dynamics in a manner consistent with the known properties of the individual channels, while voltage gating does not appear to play a role. The reaction free energy landscapes thus calculated reveal a non-trivial dependence of the αSyn/channel complex stability on the surface density of αSyn.


Subject(s)
Hemolysin Proteins , alpha-Synuclein , Anions/metabolism , Hemolysin Proteins/metabolism , Mitochondrial Membranes/metabolism , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/metabolism , alpha-Synuclein/metabolism
18.
J Am Chem Soc ; 144(32): 14564-14577, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35925797

ABSTRACT

The voltage-dependent anion channel (VDAC) is a ß-barrel channel of the mitochondrial outer membrane (MOM) that passively transports ions, metabolites, polypeptides, and single-stranded DNA. VDAC responds to a transmembrane potential by "gating," i.e. transitioning to one of a variety of low-conducting states of unknown structure. The gated state results in nearly complete suppression of multivalent mitochondrial metabolite (such as ATP and ADP) transport, while enhancing calcium transport. Voltage gating is a universal property of ß-barrel channels, but VDAC gating is anomalously sensitive to transmembrane potential. Here, we show that a single residue in the pore interior, K12, is responsible for most of VDAC's voltage sensitivity. Using the analysis of over 40 µs of atomistic molecular dynamics (MD) simulations, we explore correlations between motions of charged residues inside the VDAC pore and geometric deformations of the ß-barrel. Residue K12 is bistable; its motions between two widely separated positions along the pore axis enhance the fluctuations of the ß-barrel and augment the likelihood of gating. Single channel electrophysiology of various K12 mutants reveals a dramatic reduction of the voltage-induced gating transitions. The crystal structure of the K12E mutant at a resolution of 2.6 Å indicates a similar architecture of the K12E mutant to the wild type; however, 60 µs of atomistic MD simulations using the K12E mutant show restricted motion of residue 12, due to enhanced connectivity with neighboring residues, and diminished amplitude of barrel motions. We conclude that ß-barrel fluctuations, governed particularly by residue K12, drive VDAC gating transitions.


Subject(s)
Mitochondrial Membranes , Voltage-Dependent Anion Channels , Membrane Potentials , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Molecular Dynamics Simulation , Voltage-Dependent Anion Channels/metabolism
19.
J Membr Biol ; 255(1): 107-116, 2022 02.
Article in English | MEDLINE | ID: mdl-34731249

ABSTRACT

ERK1 is one of the members of the mitogen-activated protein kinases that regulate important cellular functions. VDAC is located at the outer membrane of mitochondria. Here, an interaction between VDAC and ERK1 has been studied on an artificial planar lipid bilayer using in vitro electrophysiology experiments. We report that VDAC is phosphorylated by ERK1 in the presence of Mg2+-ATP and its single-channel currents are inhibited on the artificial bilayer membrane. Treatment of Alkaline phosphatase on ERK1 phosphorylated VDAC leads to partial recovery of the single-channel VDAC currents. Later, phosphorylation of VDAC was demonstrated by Pro-Q diamond dye. Mass Spectrometric studies indicate phosphorylation of VDAC at Threonine 33, Threonine 55, and Serine 35. In a nutshell, phosphorylation of VDAC leads to the closure of the channel.


Subject(s)
Mitochondria , Voltage-Dependent Anion Channels , Lipid Bilayers/chemistry , Mitochondria/metabolism , Phosphorylation , Threonine/metabolism , Voltage-Dependent Anion Channels/metabolism
20.
Int J Mol Sci ; 24(1)2022 Dec 23.
Article in English | MEDLINE | ID: mdl-36613710

ABSTRACT

The voltage-dependent anion channel (VDAC) is the main passageway for ions and metabolites over the outer mitochondrial membrane. It was associated with many physiological processes, including apoptosis and modulation of intracellular Ca2+ signaling. The protein is formed by a barrel of 19 beta-sheets with an N-terminal helix lining the inner pore. Despite its large diameter, the channel can change its selectivity for ions and metabolites based on its open state to regulate transport into and out of mitochondria. VDAC was shown to be regulated by a variety of cellular factors and molecular partners including proteins, lipids and ions. Although the physiological importance of many of these modulatory effects are well described, the binding sites for molecular partners are still largely unknown. The highly symmetrical and sleek structure of the channel makes predictions of functional moieties difficult. However, one residue repeatedly sticks out when reviewing VDAC literature. A glutamate at position 73 (E73) located on the outside of the channel facing the hydrophobic membrane environment was repeatedly proposed to be involved in channel regulation on multiple levels. Here, we review the distinct hypothesized roles of E73 and summarize the open questions around this mysterious residue.


Subject(s)
Glutamic Acid , Voltage-Dependent Anion Channel 1 , Glutamic Acid/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channels/metabolism , Mitochondrial Membranes/metabolism , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL