Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 77
1.
Cancer Med ; 13(11): e7396, 2024 Jun.
Article En | MEDLINE | ID: mdl-38881325

BACKGROUND: Ovarian cancer is a common gynecological tumor with high malignant potential and poor prognosis. TRIM8, is involved in the development of various tumors, but its precise regulatory role in ovarian cancer is still unknown. AIMS: The aim of this study was to explore the specific mechanism by which TRIM8 regulates ovarian cancer. MATERIALS AND METHODS: We used bioinformatics analysis to screen for high expression of TRIM8 in ovarian cancer. The expression of TRIM8 in healthy and cancerous ovarian tissues was assessed by immunofluorescence. TRIM8 was silenced or overexpressed in ovarian cancer cell lines, with cell proliferation and migration evaluated by CCK8, transwell and clonal formation assays. The effect of TRIM8 on ovarian cancer cells in vivo was assessed by subcutaneous tumor formation experiments in nude mice. The potential interacting protein VDAC2 was identified by mass spectrometry. The mechanism underlying TRIM8 regulation of VDAC2 was evaluated by co-immunoprecipitation and western blotting. RESULTS: TRIM8 was overexpressed in ovarian cancer. TRIM8 promoted the proliferation and migration of ovarian cancer cells in vitro and the growth of subcutaneous tumors in mice in vivo. TRIM8 interacted with VDAC2, weakened the stability of the protein, and promoted its polyubiquitination and subsequent degradation. Knockdown of VDAC2 increased the resistance of ovarian cancer cells to iron death, whereas overexpression of VDAC2 attenuated ovarian cancer progression induced by TRIM8 overexpression. DISCUSSION: TRIM8 promotes ovarian cancer proliferation and migration by targeting VDAC2 for ubiquitination and degradation, these finding may provide new targets for the treatment of ovarian cancer. CONCLUSION: TRIM8 degraded VDAC2 through the ubiquitination pathway, increased the resistance of ovarian cancer cells to iron death, and promoted the proliferation and migration of ovarian cancer.


Cell Movement , Cell Proliferation , Mice, Nude , Ovarian Neoplasms , Ubiquitination , Voltage-Dependent Anion Channel 2 , Humans , Female , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Animals , Mice , Voltage-Dependent Anion Channel 2/metabolism , Voltage-Dependent Anion Channel 2/genetics , Cell Line, Tumor , Proteolysis , Gene Expression Regulation, Neoplastic , Xenograft Model Antitumor Assays
2.
Fish Shellfish Immunol ; 150: 109622, 2024 Jul.
Article En | MEDLINE | ID: mdl-38740227

The voltage-dependent anion channel 2 (VDAC2) is the abundant protein in the outer mitochondrial membrane. Opening VDAC2 pores leads to the induction of mitochondrial energy and material transport, facilitating interaction with various mitochondrial proteins implicated in essential processes such as cell apoptosis and proliferation. To investigate the VDAC2 in lower vertebrates, we identified Lr-VDAC2, a homologue of VDAC2 found in lamprey (Lethenteron reissneri), sharing a sequence identity of greater than 50 % with its counterparts. Phylogenetic analysis revealed that the position of Lr-VDAC2 aligns with the lamprey phylogeny, indicating its evolutionary relationship within the species. The Lr-VDAC2 protein was primarily located in the mitochondria of lamprey cells. The expression of the Lr-VDAC2 protein was elevated in high energy-demanding tissues, such as the gills, muscles, and myocardial tissue in normal lampreys. Lr-VDAC2 suppressed H2O2 (hydrogen peroxide)-induced 293 T cell apoptosis by reducing the expression levels of Caspase 3, Caspase 9, and Cyt C (cytochrome c). Further research into the mechanism indicated that the Lr-VDAC2 protein inhibited the pro-apoptotic activity of BAK (Bcl-2 antagonist/killer) protein by downregulating its expression at the protein translational level, thus exerting an anti-apoptotic function similar to the role of VDAC2 in humans.


Apoptosis , Down-Regulation , Fish Proteins , Hydrogen Peroxide , Lampreys , Voltage-Dependent Anion Channel 2 , bcl-2 Homologous Antagonist-Killer Protein , Animals , Voltage-Dependent Anion Channel 2/genetics , Apoptosis/drug effects , Lampreys/genetics , Lampreys/immunology , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Humans , Down-Regulation/drug effects , Fish Proteins/genetics , Fish Proteins/immunology , HEK293 Cells , Gene Expression Regulation/drug effects , Phylogeny , Sequence Alignment/veterinary , Amino Acid Sequence , Gene Expression Profiling/veterinary
3.
PLoS Biol ; 22(5): e3002617, 2024 May.
Article En | MEDLINE | ID: mdl-38696533

BAK and BAX execute intrinsic apoptosis by permeabilising the mitochondrial outer membrane. Their activity is regulated through interactions with pro-survival BCL-2 family proteins and with non-BCL-2 proteins including the mitochondrial channel protein VDAC2. VDAC2 is important for bringing both BAK and BAX to mitochondria where they execute their apoptotic function. Despite this important function in apoptosis, while interactions with pro-survival family members are well characterised and have culminated in the development of drugs that target these interfaces to induce cancer cell apoptosis, the interaction between BAK and VDAC2 remains largely undefined. Deep scanning mutagenesis coupled with cysteine linkage identified key residues in the interaction between BAK and VDAC2. Obstructive labelling of specific residues in the BH3 domain or hydrophobic groove of BAK disrupted this interaction. Conversely, mutating specific residues in a cytosol-exposed region of VDAC2 stabilised the interaction with BAK and inhibited BAK apoptotic activity. Thus, this VDAC2-BAK interaction site can potentially be targeted to either inhibit BAK-mediated apoptosis in scenarios where excessive apoptosis contributes to disease or to promote BAK-mediated apoptosis for cancer therapy.


Apoptosis , Voltage-Dependent Anion Channel 2 , bcl-2 Homologous Antagonist-Killer Protein , Voltage-Dependent Anion Channel 2/metabolism , Voltage-Dependent Anion Channel 2/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , Humans , Protein Binding , Mitochondria/metabolism , Animals , HEK293 Cells
4.
Insect Biochem Mol Biol ; 169: 104125, 2024 Jun.
Article En | MEDLINE | ID: mdl-38616030

Voltage-dependent anion channel 2 (VDAC2) is an important channel protein that plays a crucial role in the host response to viral infection. The receptor for activated C kinase 1 (RACK1) is also a key host factor involved in viral replication. Our previous research revealed that Bombyx mori VDAC2 (BmVDAC2) and B. mori RACK1 (BmRACK1) may interact with Bombyx mori nucleopolyhedrovirus (BmNPV), though the specific molecular mechanism remains unclear. In this study, the interaction between BmVDAC2 and BmRACK1 in the mitochondria was determined by various methods. We found that BmNPV p35 interacts directly with BmVDAC2 rather than BmRACK1. BmNPV infection significantly reduced the expression of BmVDAC2, and activated the mitochondrial apoptosis pathway. Overexpression of BmVDAC2 in BmN cells inhibited BmNPV-induced cytochrome c (cyto c) release, decrease in mitochondrial membrane potential as well as apoptosis. Additionally, the inhibition of cyto c release by BmVDAC2 requires the involvement of BmRACK1 and protein kinase C. Interestingly, overexpression of p35 inhibited cyto c release during mitochondrial apoptosis in a RACK1 and VDAC2-dependent manner. Even the mutant p35, which loses Caspase inhibitory activity, could still bind to VDAC2 and inhibit cyto c release. In summary, our results indicated that BmNPV p35 interacts with the VDAC2-RACK1 complex to regulate apoptosis by inhibiting cyto c release. These findings confirm the interaction between BmVDAC2 and BmRACK1, the interaction between p35 and the VDAC2-RACK1 complex, and a novel target that BmNPV p35 regulates apoptosis in Bombyx mori via interaction with the BmVDAC2-BmRACK1 complex. The result provide an initial exploration of the function of this interaction in the BmNPV-induced mitochondrial apoptosis pathway.


Apoptosis , Bombyx , Insect Proteins , Nucleopolyhedroviruses , Receptors for Activated C Kinase , Animals , Bombyx/virology , Bombyx/metabolism , Bombyx/genetics , Nucleopolyhedroviruses/physiology , Receptors for Activated C Kinase/metabolism , Receptors for Activated C Kinase/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , Voltage-Dependent Anion Channel 2/metabolism , Voltage-Dependent Anion Channel 2/genetics , Mitochondria/metabolism
5.
Aging (Albany NY) ; 16(4): 3160-3184, 2024 Feb 20.
Article En | MEDLINE | ID: mdl-38382091

Non-small cell lung cancer (NSCLC) is characterized by stronger metastatic ability and worse prognosis. In NSCLC, hypoxia is a major cause of invasion and metastasis through promoting angiogenesis. In present study, NSCLC cell clusters were extracted from single cell-sequencing dataset GSE131907, which were combined with hypoxia-related genes to group clusters. qRT-PCR and western blot were used to validate the expression of target gene. Nine NSCLC clusters were extracted, which were divided into two hypoxia-related subgroups, C1 and C2. Totally 101 differentially expressed prognostic genes were identified between subgroups. Of which, VDAC2 showed excellent prognostic value for NSCLC and was selected for further analysis. VDAC2 was upregulated in tumor samples in TCGA and was correlated with advanced stages. In vitro experiments validated this trend. Five crucial immune cells showed differential infiltration proportions between high and low VDAC2 expression groups. VDAC2 knockdown significantly inhibited the proliferation and invasion ability of NSCLC cells. Integrating single cell and bulk sequencing data as well as wet lab experiments, hypoxia-related VDAC2 exhibited important prognostic value and showed the promise of becoming immune-therapy target in NSCLC.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Cell Line, Tumor , Prognosis , MicroRNAs/genetics , Sequence Analysis, RNA , Hypoxia , Voltage-Dependent Anion Channel 2/genetics
6.
Int J Biol Sci ; 19(10): 3143-3158, 2023.
Article En | MEDLINE | ID: mdl-37416771

Sepsis-induced myocardial dysfunction (SIMD) is a prevalent and severe form of organ dysfunction with elusive underlying mechanisms and limited treatment options. In this study, the cecal ligation and puncture and lipopolysaccharide (LPS) were used to reproduce sepsis model in vitro and vivo. The level of voltage-dependent anion channel 2 (VDAC2) malonylation and myocardial malonyl-CoA were detected by mass spectrometry and LC-MS-based metabolomics. Role of VDAC2 malonylation on cardiomyocytes ferroptosis and treatment effect of mitochondrial targeting nano material TPP-AAV were observed. The results showed that VDAC2 lysine malonylation was significantly elevated after sepsis. In addition, the regulation of VDAC2 lysine 46 (K46) malonylation by K46E and K46Q mutation affected mitochondrial-related ferroptosis and myocardial injury. The molecular dynamic simulation and circular dichroism further demonstrated that VDAC2 malonylation altered the N-terminus structure of the VDAC2 channel, causing mitochondrial dysfunction, increasing mitochondrial ROS levels, and leading to ferroptosis. Malonyl-CoA was identified as the primary inducer of VDAC2 malonylation. Furthermore, the inhibition of malonyl-CoA using ND-630 or ACC2 knock-down significantly reduced the malonylation of VDAC2, decreased the occurrence of ferroptosis in cardiomyocytes, and alleviated SIMD. The study also found that the inhibition of VDAC2 malonylation by synthesizing mitochondria targeting nano material TPP-AAV could further alleviate ferroptosis and myocardial dysfunction following sepsis. In summary, our findings indicated that VDAC2 malonylation plays a crucial role in SIMD and that targeting VDAC2 malonylation could be a potential treatment strategy for SIMD.


Ferroptosis , Sepsis , Humans , Voltage-Dependent Anion Channel 2/genetics , Lysine , Mitochondria , Sepsis/complications
7.
Hepatology ; 75(2): 403-418, 2022 02.
Article En | MEDLINE | ID: mdl-34435375

BACKGROUND AND AIMS: Although the prevalence of NAFLD has risen dramatically to 25% of the adult population worldwide, there are as yet no approved pharmacological interventions for the disease because of uncertainty about the underlying molecular mechanisms. It is known that mitochondrial dysfunction is an important factor in the development of NAFLD. Mitochondrial antiviral signaling protein (MAVS) is a critical signaling adaptor for host defenses against viral infection. However, the role of MAVS in mitochondrial metabolism during NAFLD progression remains largely unknown. APPROACH AND RESULTS: Based on expression analysis, we identified a marked down-regulation of MAVS in hepatocytes during NAFLD progression. By using MAVS global knockout and hepatocyte-specific MAVS knockout mice, we found that MAVS is protective against diet-induced NAFLD. MAVS deficiency induces extensive mitochondrial dysfunction during NAFLD pathogenesis, which was confirmed as impaired mitochondrial respiratory capacity and membrane potential. Metabolomics data also showed the extensive metabolic disorders after MAVS deletion. Mechanistically, MAVS interacts with the N-terminal stretch of voltage-dependent anion channel 2 (VDAC2), which is required for the ability of MAVS to influence mitochondrial function and hepatic steatosis. CONCLUSIONS: In hepatocytes, MAVS plays an important role in protecting against NAFLD by helping to regulate healthy mitochondrial function. These findings provide insights regarding the metabolic importance of conventional immune regulators and support the possibility that targeting MAVS may represent an avenue for treating NAFLD.


Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Animals , Cells, Cultured , Disease Progression , Down-Regulation , Gene Knockdown Techniques , Hepatic Stellate Cells , Hepatocytes , Homeostasis , Humans , Lipogenesis/genetics , Male , Metabolomics , Mice , Mice, Knockout , Mitochondria/physiology , Non-alcoholic Fatty Liver Disease/genetics , Primary Cell Culture , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism
8.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article En | MEDLINE | ID: mdl-34446558

Calcineurin is a calcium-dependent phosphatase that plays roles in a variety of biological processes including immune responses. In spermatozoa, there is a testis-enriched calcineurin composed of PPP3CC and PPP3R2 (sperm calcineurin) that is essential for sperm motility and male fertility. Because sperm calcineurin has been proposed as a target for reversible male contraceptives, identifying proteins that interact with sperm calcineurin widens the choice for developing specific inhibitors. Here, by screening the calcineurin-interacting PxIxIT consensus motif in silico and analyzing the function of candidate proteins through the generation of gene-modified mice, we discovered that SPATA33 interacts with sperm calcineurin via a PQIIIT sequence. Spata33 knockout mice exhibit reduced sperm motility because of an inflexible midpiece, leading to impaired male fertility, which phenocopies Ppp3cc and Ppp3r2 knockout mice. Further analysis reveals that sperm calcineurin disappears from the mitochondria in the Spata33 knockout testis. In addition, immunoprecipitation analysis indicates that sperm calcineurin interacts with not only SPATA33 but also the mitochondrial protein VDAC2. These results indicate that SPATA33 localizes calcineurin to the mitochondria and regulates sperm motility.


Calcineurin/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Mitochondria/metabolism , Sperm Motility , Testis/physiology , Voltage-Dependent Anion Channel 2/metabolism , Animals , Calcineurin/genetics , Female , Male , Mice , Mice, Knockout , Spermatogenesis , Voltage-Dependent Anion Channel 2/genetics
9.
Nat Commun ; 12(1): 4583, 2021 07 28.
Article En | MEDLINE | ID: mdl-34321484

Voltage dependent anion channel 2 (VDAC2) is an outer mitochondrial membrane porin known to play a significant role in apoptosis and calcium signaling. Abnormalities in calcium homeostasis often leads to electrical and contractile dysfunction and can cause dilated cardiomyopathy and heart failure. However, the specific role of VDAC2 in intracellular calcium dynamics and cardiac function is not well understood. To elucidate the role of VDAC2 in calcium homeostasis, we generated a cardiac ventricular myocyte-specific developmental deletion of Vdac2 in mice. Our results indicate that loss of VDAC2 in the myocardium causes severe impairment in excitation-contraction coupling by altering both intracellular and mitochondrial calcium signaling. We also observed adverse cardiac remodeling which progressed to severe cardiomyopathy and death. Reintroduction of VDAC2 in 6-week-old knock-out mice partially rescued the cardiomyopathy phenotype. Activation of VDAC2 by efsevin increased cardiac contractile force in a mouse model of pressure-overload induced heart failure. In conclusion, our findings demonstrate that VDAC2 plays a crucial role in cardiac function by influencing cellular calcium signaling. Through this unique role in cellular calcium dynamics and excitation-contraction coupling VDAC2 emerges as a plausible therapeutic target for heart failure.


Calcium/metabolism , Cardiomyopathy, Dilated/metabolism , Homeostasis , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism , Animals , Apoptosis , Calcium Signaling , Cardiomyopathy, Dilated/mortality , Heart Failure/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Transcriptome
10.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article En | MEDLINE | ID: mdl-33536340

The mammalian sperm midpiece has a unique double-helical structure called the mitochondrial sheath that wraps tightly around the axoneme. Despite the remarkable organization of the mitochondrial sheath, the molecular mechanisms involved in mitochondrial sheath formation are unclear. In the process of screening testis-enriched genes for functions in mice, we identified armadillo repeat-containing 12 (ARMC12) as an essential protein for mitochondrial sheath formation. Here, we engineered Armc12-null mice, FLAG-tagged Armc12 knock-in mice, and TBC1 domain family member 21 (Tbc1d21)-null mice to define the functions of ARMC12 in mitochondrial sheath formation in vivo. We discovered that absence of ARMC12 causes abnormal mitochondrial coiling along the flagellum, resulting in reduced sperm motility and male sterility. During spermiogenesis, sperm mitochondria in Armc12-null mice cannot elongate properly at the mitochondrial interlocking step which disrupts abnormal mitochondrial coiling. ARMC12 is a mitochondrial peripheral membrane protein and functions as an adherence factor between mitochondria in cultured cells. ARMC12 in testicular germ cells interacts with mitochondrial proteins MIC60, VDAC2, and VDAC3 as well as TBC1D21 and GK2, which are required for mitochondrial sheath formation. We also observed that TBC1D21 is essential for the interaction between ARMC12 and VDAC proteins in vivo. These results indicate that ARMC12 uses integral mitochondrial membrane proteins VDAC2 and VDAC3 as scaffolds to link mitochondria and works cooperatively with TBC1D21. Thus, our studies have revealed that ARMC12 regulates spatiotemporal mitochondrial dynamics to form the mitochondrial sheath through cooperative interactions with several proteins on the sperm mitochondrial surface.


Armadillo Domain Proteins/genetics , GTPase-Activating Proteins/genetics , Infertility, Male/genetics , Microfilament Proteins/genetics , Mitochondrial Dynamics/genetics , Animals , Axoneme/genetics , Humans , Infertility, Male/pathology , Male , Mice , Mice, Knockout , Mitochondrial Membrane Transport Proteins/genetics , Sperm Motility/genetics , Sperm Tail/pathology , Sperm Tail/ultrastructure , Spermatids/metabolism , Spermatogenesis/genetics , Spermatozoa/pathology , Spermatozoa/ultrastructure , Testis/metabolism , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channels/genetics
11.
Cell Death Differ ; 28(3): 1076-1090, 2021 03.
Article En | MEDLINE | ID: mdl-33087875

Selective autophagic degradation of mitochondria (mitophagy) is important in maintaining proper cellular homeostasis. Here, we found that SPATA33 is a novel autophagy mediator for mitophagy in testis. The SPATA33 protein localizes on mitochondria via its binding of the carboxyl terminal with the outer mitochondrial membrane protein VDAC2. Upon starvation induction, SPATA33 is recruited to autophagosome by binding the autophagy machinery ATG16L1 via its N-terminal along with mitochondria. Notably, Spata33 knockout inhibited autophagy and overexpression can promote autophagosome formation for mitochondrial sequestration. Therefore, SPATA33 confers selectivity for mitochondrial degradation and promotes mitophagy in male germline cells.


Autophagosomes/metabolism , Autophagy-Related Proteins/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Mitochondria/metabolism , Mitophagy/physiology , Voltage-Dependent Anion Channel 2/metabolism , Animals , Autophagy-Related Proteins/genetics , Cell Line , Germ Cells , Humans , Male , Mice , Mice, Inbred ICR , Mitochondria/pathology , Testis/physiology , Voltage-Dependent Anion Channel 2/genetics
12.
J Cell Sci ; 133(21)2020 11 10.
Article En | MEDLINE | ID: mdl-33067255

Cytoskeleton-associated protein 4 (CKAP4) is a palmitoylated type II transmembrane protein localized to the endoplasmic reticulum (ER). Here, we found that knockout (KO) of CKAP4 in HeLaS3 cells induces the alteration of mitochondrial structures and increases the number of ER-mitochondria contact sites. To understand the involvement of CKAP4 in mitochondrial functions, the binding proteins of CKAP4 were explored, enabling identification of the mitochondrial porin voltage-dependent anion-selective channel protein 2 (VDAC2), which is localized to the outer mitochondrial membrane. Palmitoylation at Cys100 of CKAP4 was required for the binding between CKAP4 and VDAC2. In CKAP4 KO cells, the binding of inositol trisphosphate receptor (IP3R) and VDAC2 was enhanced, the intramitochondrial Ca2+ concentration increased and the mitochondrial membrane potential decreased. In addition, CKAP4 KO decreased the oxidative consumption rate, in vitro cancer cell proliferation under low-glucose conditions and in vivo xenograft tumor formation. The phenotypes were not rescued by expression of a palmitoylation-deficient CKAP4 mutant. These results suggest that CKAP4 plays a role in maintaining mitochondrial functions through the binding to VDAC2 at ER-mitochondria contact sites and that palmitoylation is required for this novel function of CKAP4.This article has an associated First Person interview with the first author of the paper.


Endoplasmic Reticulum , Membrane Proteins/genetics , Mitochondria , Voltage-Dependent Anion Channel 2/genetics , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Lipoylation , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism
13.
Sci Rep ; 10(1): 16751, 2020 10 12.
Article En | MEDLINE | ID: mdl-33046783

Bisindolylpyrrole at 0.1 µM is cytoprotective in 2% FBS that is counteracted by cyclosporin-A (CsA), an inhibitor of cyclophilin-D (CypD). We hypothesized that the cytoprotective effect might be due to transient mitochondrial permeability transition (tPT). This study tested the hypothesis that bisindolylpyrrole can trigger tPT extensively, thereby leading to cell death under certain conditions. Indeed, CsA-sensitive tPT-mediated apoptosis could be induced by bisindolylpyrrole at > 5 µM in HeLa cells cultured in 0.1% FBS, depending on CypD and VDAC1/2, as shown by siRNA knockdown experiments. Rat liver mitochondria also underwent swelling in response to bisindolylpyrrole, which proceeded at a slower rate than Ca2+-induced swelling, and which was blocked by the VDAC inhibitor tubulin and the ANT inhibitor bongkrekate, indicating the involvement of the ANT-associated, smaller pore. We examined why 0.1% FBS is a prerequisite for apoptosis and found that apoptosis is blocked by PKC activation, which is counteracted by the overexpressed defective PKCε. In mitochondrial suspensions, bisindolylpyrrole triggered CsA-sensitive swelling, which was suppressed selectively by pretreatment with PKCε, but not in the co-presence of tubulin. These data suggest that upon PKC inactivation the cytoprotective compound bisindolylpyrrole can induce prolonged tPT causing apoptosis in a CypD-dependent manner through the VDAC1/2-regulated ANT-associated pore.


Apoptosis/drug effects , Apoptosis/genetics , Cytoprotection/drug effects , Cytoprotection/genetics , Mitochondria, Liver/metabolism , Mitochondrial ADP, ATP Translocases/genetics , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Transmembrane Permeability-Driven Necrosis/drug effects , Peptidyl-Prolyl Isomerase F/genetics , Peptidyl-Prolyl Isomerase F/metabolism , Pyrroles/pharmacology , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism , Adenosine Diphosphate , Animals , Calcium/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Protein Kinase C/metabolism , Protein Kinase C/physiology , RNA, Small Interfering/genetics , Rats
14.
J Biol Chem ; 295(43): 14653-14665, 2020 10 23.
Article En | MEDLINE | ID: mdl-32817169

Transmembrane ß-barrels of eukaryotic outer mitochondrial membranes (OMMs) are major channels of communication between the cytosol and mitochondria and are indispensable for cellular homeostasis. A structurally intriguing exception to all known transmembrane ß-barrels is the unique odd-stranded, i.e. 19-stranded, structures found solely in the OMM. The molecular origins of this 19-stranded structure and its associated functional significance are unclear. In humans, the most abundant OMM transporter is the voltage-dependent anion channel. Here, using the human voltage-dependent anion channel as our template scaffold, we designed and engineered odd- and even-stranded structures of smaller (V216, V217, V218) and larger (V220, V221) barrel diameters. Determination of the structure, dynamics, and energetics of these engineered structures in bilayer membranes reveals that the 19-stranded barrel surprisingly holds modest to low stability in a lipid-dependent manner. However, we demonstrate that this structurally metastable protein possesses superior voltage-gated channel regulation, efficient mitochondrial targeting, and in vivo cell survival, with lipid-modulated stability, all of which supersede the occurrence of a metastable 19-stranded scaffold. We propose that the unique structural adaptation of these transmembrane transporters exclusively in mitochondria bears strong evolutionary basis and is functionally significant for homeostasis.


Lipid Bilayers/metabolism , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/metabolism , Animals , Evolution, Molecular , Humans , Lipid Bilayers/chemistry , Mitochondria/chemistry , Mitochondria/genetics , Mitochondria/metabolism , Models, Molecular , Mutation , Porins/chemistry , Porins/genetics , Porins/metabolism , Protein Conformation, beta-Strand , Protein Engineering , Protein Stability , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Thermodynamics , Voltage-Dependent Anion Channel 2/chemistry , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channel 2/metabolism , Voltage-Dependent Anion Channels/genetics
15.
Tumour Biol ; 42(8): 1010428320951057, 2020 Aug.
Article En | MEDLINE | ID: mdl-32829673

The exchange of metabolites between mitochondria and cytosol occurs through pores formed by voltage-dependent anion channel proteins. Voltage-dependent anion channels appear to be master regulators of mitochondrial bioenergetics and the intracellular flow of energy. Deregulation of voltage-dependent anion channels expression is thought to be related to mitochondrial dysfunction in cancer. The aim of this study was to investigate the mRNA and protein expression levels of VDAC1, VDAC2, and VDAC3 in relation to clinicopathological characteristics of endometrial cancer as well as the prognostic significance of voltage-dependent anion channels expression for overall survival. VDAC1 and VDAC3 expressions were significantly higher in cancer compared to normal tissues. Kaplan-Meier analysis indicated that high expression of all VDAC genes or high VDAC2 protein level predicted poor overall survival. Multivariate analysis identified the VDAC1 and VDAC2 mRNA levels as well as VDAC2 protein level as independent prognostic factors. Our results suggest that increased expression of voltage-dependent anion channels correlates with tumor progression and may serve as a potential prognostic biomarker in endometrial cancer.


Endometrial Neoplasms/pathology , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/genetics , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channels/genetics , Amino Acid Sequence , Biomarkers, Tumor/genetics , Cytoplasm/metabolism , Endometrial Neoplasms/mortality , Female , Humans , Middle Aged , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/biosynthesis , Prognosis , RNA, Messenger/genetics , Voltage-Dependent Anion Channel 1/biosynthesis , Voltage-Dependent Anion Channel 2/biosynthesis , Voltage-Dependent Anion Channels/biosynthesis
16.
Theranostics ; 10(16): 7178-7192, 2020.
Article En | MEDLINE | ID: mdl-32641986

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide and effective therapy remains a challenge. IFIT3 is an interferon-stimulated gene with antiviral and pro-inflammatory functions. Our previous work has shown that high expression of IFIT3 is correlated with poor survival in PDAC patients who receive chemotherapy suggesting a link between IFIT3 and chemotherapy resistance in PDAC. However, the exact role and molecular mechanism of IFIT3 in chemotherapy resistance in PDAC has been unclear. Methods: A group of transcriptome datasets were downloaded and analyzed for the characterization of IFIT3 in PDAC. Highly metastatic PDAC cell line L3.6pl and patient-derived primary cell TBO368 were used and IFIT3 knockdown and the corresponding knockin cells were established for in vitro studies. Chemotherapy-induced apoptosis, ROS production, confocal immunofluorescence, subcellular fractionation, chromatin-immunoprecipitation, co-immunoprecipitation and mass spectrometry analysis were determined to further explore the biological role of IFIT3 in chemotherapy resistance of PDAC. Results: Based on PDAC transcriptome data, we show that IFIT3 expression is associated with the squamous molecular subtype of PDAC and an increase in inflammatory response and apoptosis pathways. We further identify a crucial role for IFIT3 in the regulation of mitochondria-associated apoptosis during chemotherapy. Knockdown of IFIT3 attenuates the chemotherapy resistance of PDAC cells to gemcitabine, paclitaxel, and FOLFIRINOX regimen treatments, independent of individual chemotherapy regimens. While IFIT3 overexpression was found to promote drug resistance. Co-immunoprecipitation identified a direct interaction between IFIT3 and the mitochondrial channel protein VDAC2, an important regulator of mitochondria-associated apoptosis. It was subsequently found that IFIT3 regulates the post-translational modification-O-GlcNAcylation of VDAC2 by stabilizing the interaction of VDAC2 with O-GlcNAc transferase. Increased O-GlcNAcylation of VDAC2 protected PDAC cells from chemotherapy induced apoptosis. Conclusions: These results effectively demonstrate a central mechanism by which IFIT3 expression can affect chemotherapy resistance in PDAC. Targeting IFIT3/VDAC2 may represent a novel strategy to sensitize aggressive forms of pancreatic cancer to conventional chemotherapy regimens.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Voltage-Dependent Anion Channel 2/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/genetics , Apoptosis/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Cell Proliferation/genetics , Datasets as Topic , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/immunology , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Kaplan-Meier Estimate , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/mortality , Primary Cell Culture , Prognosis , Protein Processing, Post-Translational/immunology , RNA-Seq
17.
Nat Commun ; 11(1): 433, 2020 01 23.
Article En | MEDLINE | ID: mdl-31974380

Ferroptosis is a newly defined form of regulated cell death characterized by the iron-dependent accumulation of lipid hydroperoxides. Erastin, the ferroptosis activator, binds to voltage-dependent anion channels VDAC2 and VDCA3, but treatment with erastin can result in the degradation of the channels. Here, the authors show that Nedd4 is induced following erastin treatment, which leads to the ubiquitination and subsequent degradation of the channels. Depletion of Nedd4 limits the protein degradation of VDAC2/3, which increases the sensitivity of cancer cells to erastin. By understanding the molecular mechanism of erastin-induced cellular resistance, we can discover how cells adapt to new molecules to maintain homeostasis. Furthermore, erastin-induced resistance mediated by FOXM1-Nedd4-VDAC2/3 negative feedback loop provides an initial framework for creating avenues to overcome the drug resistance of ferroptosis activators.


Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Mitochondrial Membrane Transport Proteins/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Piperazines/pharmacology , Voltage-Dependent Anion Channel 2/metabolism , Voltage-Dependent Anion Channels/metabolism , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Female , Ferroptosis/drug effects , Ferroptosis/physiology , Forkhead Box Protein M1/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , Mice, Nude , Mitochondrial Membrane Transport Proteins/genetics , Nedd4 Ubiquitin Protein Ligases/genetics , Ubiquitination/drug effects , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channels/genetics , Xenograft Model Antitumor Assays
18.
mBio ; 10(4)2019 08 20.
Article En | MEDLINE | ID: mdl-31431557

Microsporidia are opportunistic intracellular pathogens that can infect a wide variety of hosts ranging from invertebrates to vertebrates. During invasion, the microsporidian polar tube pushes into the host cell, creating a protective microenvironment, the invasion synapse, into which the sporoplasm extrudes. Within the synapse, the sporoplasm then invades the host cell, forming a parasitophorous vacuole (PV). Using a proteomic approach, we identified Encephalitozoon hellem sporoplasm surface protein 1 (EhSSP1), which localized to the surface of extruded sporoplasms. EhSSP1 was also found to interact with polar tube protein 4 (PTP4). Recombinant EhSSP1 (rEhSSP1) bound to human foreskin fibroblasts, and both anti-EhSSP1 and rEhSSP1 caused decreased levels of host cell invasion, suggesting that interaction of SSP1 with the host cell was involved in invasion. Coimmunoprecipitation (Co-IP) followed by proteomic analysis identified host cell voltage-dependent anion channels (VDACs) as EhSSP1 interacting proteins. Yeast two-hybrid assays demonstrated that EhSSP1 was able to interact with VDAC1, VDAC2, and VDAC3. rEhSSP1 colocalized with the host mitochondria which were associated with microsporidian PVs in infected cells. Transmission electron microscopy revealed that the outer mitochondrial membrane interacted with meronts and the PV membrane, mitochondria clustered around meronts, and the VDACs were concentrated at the interface of mitochondria and parasite. Knockdown of VDAC1, VDAC2, and VDAC3 in host cells resulted in significant decreases in the number and size of the PVs and a decrease in mitochondrial PV association. The interaction of EhSSP1 with VDAC probably plays an important part in energy acquisition by microsporidia via its role in the association of mitochondria with the PV.IMPORTANCE Microsporidia are important opportunistic human pathogens in immune-suppressed individuals, such as those with HIV/AIDS and recipients of organ transplants. The sporoplasm is critical for establishing microsporidian infection. Despite the biological importance of this structure for transmission, there is limited information about its structure and composition that could be targeted for therapeutic intervention. Here, we identified a novel E. hellem sporoplasm surface protein, EhSSP1, and demonstrated that it can bind to host cell mitochondria via host VDAC. Our data strongly suggest that the interaction between SSP1 and VDAC is important for the association of mitochondria with the parasitophorous vacuole during microsporidian infection. In addition, binding of SSP1 to the host cell is associated with the final steps of invasion in the invasion synapse.


Host-Pathogen Interactions/physiology , Microsporidia/metabolism , Mitochondria/microbiology , Voltage-Dependent Anion Channels/metabolism , Cytoplasm , Encephalitozoon , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Knockdown Techniques , Gene Silencing , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Proteomics , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 2/genetics , Voltage-Dependent Anion Channels/genetics
19.
Fish Shellfish Immunol ; 92: 889-896, 2019 Sep.
Article En | MEDLINE | ID: mdl-31299465

Ranaviruses belong to the family Iridoviridae, and have become a serious threat to both farmed and natural populations of fish and amphibians. Previous reports showed that ranaviruses could encode viral Bcl-2 family-like proteins (vBcl-2), which play a critical role in the regulation of cell apoptosis. However, the mechanism of ranaviruses vBcl-2 interactions with host protein in mediating apoptosis remains unknown. Tiger frog virus (TFV) belonging to the genus Ranavirus has been isolated from infected tadpoles of Rana tigrina rugulosa, and it causes a high mortality rate among tiger frog tadpoles cultured in southern China. This study elucidated the molecular mechanism underlying the interaction of TFV ORF104R with the VDAC2 protein to regulate cell apoptosis. TFV ORF104R is highly similar to other ranaviruses vBcl-2 and host Mcl-1 proteins, indicating that TFV ORF104R is a postulate vBcl-2 protein. Transcription and protein expression levels showed that TFV orf104r was a late viral gene. Western blot results suggested that TFV ORF104R was a viral structural protein. Subcellular localization analysis indicated that TFV ORF104R was predominantly colocalized with the mitochondria. Overexpressed TFV ORF104R could suppress the release of cytochrome C and the activities of caspase-9 and caspase-3. These results indicated that TFV ORF104R might play an important role in anti-apoptosis. Furthermore, the interaction between TFV ORF104R and VDAC2 was detected by co-immunoprecipitation in vitro. The above observations suggest that the molecular mechanism of TFV-regulated anti-apoptosis is through the interaction of TFV ORF104R with the VDAC2 protein. Our study provided a mechanistic basis for the ranaviruses vBcl-2-mediated inhibition of apoptosis and improved the understanding on how TFV subverts host defense mechanisms in vivo.


Apoptosis/immunology , Cyprinidae , DNA Virus Infections/veterinary , Fish Diseases/immunology , Genes, Viral , Ranavirus/physiology , Voltage-Dependent Anion Channel 2/immunology , Animals , DNA Virus Infections/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Open Reading Frames , Voltage-Dependent Anion Channel 2/genetics
20.
Exp Cell Res ; 381(2): 223-234, 2019 08 15.
Article En | MEDLINE | ID: mdl-31102594

We previously reported that the Bcl-2 family member human Bcl-rambo, also known as BCL2L13, induces apoptosis in human embryonic kidney 293T cells. Mouse Bcl-rambo has recently been reported to mediate mitochondrial fragmentation and mitophagy. In the present study, we showed that the transfection of human Bcl-rambo and its microtubule-associated protein light chain 3-interacting region motif mutant (W276A/I279A) caused mitochondrial fragmentation and the perinuclear accumulation of fragmented mitochondria in human lung adenocarcinoma A549 cells. In comprehensive screening using the Drosophila model in which human Bcl-rambo was ectopically expressed in eye imaginal discs, voltage-dependent anion channels (VDAC), also known as mitochondrial porin, were found to manifest a genetic interaction with human Bcl-rambo. In addition to human adenine nucleotide translocase (ANT) 1 and ANT2, the human Bcl-rambo protein bound to human VDAC1, albeit to a lesser extent than ANT2. Moreover, human VDAC1 and human VDAC2 in particular promoted the activation of effector caspases only when they were co-expressed with human Bcl-rambo in 293T cells. Bcl-rambo induced the perinuclear accumulation of fragmented mitochondria by the knockdown of VDAC1, VDAC2, and VDAC3 in A549 cells. Thus, the present study revealed that human Bcl-rambo and VDAC cooperatively promote the activation of effector caspases in human cultured cells.


Caspases, Effector/metabolism , Drosophila/genetics , Epistasis, Genetic/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Voltage-Dependent Anion Channels/genetics , A549 Cells , Animals , Animals, Genetically Modified , Cells, Cultured , Enzyme Activation/genetics , HEK293 Cells , Humans , Mice , Mitochondrial Membrane Transport Proteins/genetics , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 2/genetics
...