162 results on '"Shteinfer-Kuzmine A"'
Search Results
2. Interaction of SMAC with a survivin-derived peptide alters essential cancer hallmarks: Tumor growth, inflammation, and immunosuppression
- Author
-
Santhanam, Manikandan, Kumar Pandey, Swaroop, Shteinfer-Kuzmine, Anna, Paul, Avijit, Abusiam, Nur, Zalk, Ran, and Shoshan-Barmatz, Varda
- Published
- 2024
- Full Text
- View/download PDF
3. Decoding Cancer through Silencing the Mitochondrial Gatekeeper VDAC1
- Author
-
Tasleem Arif, Anna Shteinfer-Kuzmine, and Varda Shoshan-Barmatz
- Subjects
siRNA ,metabolism ,mitochondria ,stem cells ,VDAC1 ,Microbiology ,QR1-502 - Abstract
Mitochondria serve as central hubs for regulating numerous cellular processes that include metabolism, apoptosis, cell cycle progression, proliferation, differentiation, epigenetics, immune signaling, and aging. The voltage-dependent anion channel 1 (VDAC1) functions as a crucial mitochondrial gatekeeper, controlling the flow of ions, such as Ca2+, nucleotides, and metabolites across the outer mitochondrial membrane, and is also integral to mitochondria-mediated apoptosis. VDAC1 functions in regulating ATP production, Ca2+ homeostasis, and apoptosis, which are essential for maintaining mitochondrial function and overall cellular health. Most cancer cells undergo metabolic reprogramming, often referred to as the “Warburg effect”, supplying tumors with energy and precursors for the biosynthesis of nucleic acids, phospholipids, fatty acids, cholesterol, and porphyrins. Given its multifunctional nature and overexpression in many cancers, VDAC1 presents an attractive target for therapeutic intervention. Our research has demonstrated that silencing VDAC1 expression using specific siRNA in various tumor types leads to a metabolic rewiring of the malignant cancer phenotype. This results in a reversal of oncogenic properties that include reduced tumor growth, invasiveness, stemness, epithelial–mesenchymal transition. Additionally, VDAC1 depletion alters the tumor microenvironment by reducing angiogenesis and modifying the expression of extracellular matrix- and structure-related genes, such as collagens and glycoproteins. Furthermore, VDAC1 depletion affects several epigenetic-related enzymes and substrates, including the acetylation-related enzymes SIRT1, SIRT6, and HDAC2, which in turn modify the acetylation and methylation profiles of histone 3 and histone 4. These epigenetic changes can explain the altered expression levels of approximately 4000 genes that are associated with reversing cancer cells oncogenic properties. Given VDAC1’s critical role in regulating metabolic and energy processes, targeting it offers a promising strategy for anti-cancer therapy. We also highlight the role of VDAC1 expression in various disease pathologies, including cardiovascular, neurodegenerative, and viral and bacterial infections, as explored through siRNA targeting VDAC1. Thus, this review underscores the potential of targeting VDAC1 as a strategy for addressing high-energy-demand cancers. By thoroughly understanding VDAC1’s diverse roles in metabolism, energy regulation, mitochondrial functions, and other cellular processes, silencing VDAC1 emerges as a novel and strategic approach to combat cancer.
- Published
- 2024
- Full Text
- View/download PDF
4. VDAC1-Based Peptides as Potential Modulators of VDAC1 Interactions with Its Partners and as a Therapeutic for Cancer, NASH, and Diabetes
- Author
-
Anna Shteinfer-Kuzmine, Manikandan Santhanam, and Varda Shoshan-Barmatz
- Subjects
apoptosis ,cancer ,mitochondria ,peptide ,VDAC1 ,Microbiology ,QR1-502 - Abstract
This review presents current knowledge related to the voltage-dependent anion channel-1 (VDAC1) as a multi-functional mitochondrial protein that acts in regulating both cell life and death. The location of VDAC1 at the outer mitochondrial membrane (OMM) allows control of metabolic cross-talk between the mitochondria and the rest of the cell, and also enables its interaction with proteins that are involved in metabolic, cell death, and survival pathways. VDAC1′s interactions with over 150 proteins can mediate and regulate the integration of mitochondrial functions with cellular activities. To target these protein–protein interactions, VDAC1-derived peptides have been developed. This review focuses specifically on cell-penetrating VDAC1-based peptides that were developed and used as a “decoy” to compete with VDAC1 for its VDAC1-interacting proteins. These peptides interfere with VDAC1 interactions, for example, with metabolism-associated proteins such as hexokinase (HK), or with anti-apoptotic proteins such as Bcl-2 and Bcl-xL. These and other VDAC1-interacting proteins are highly expressed in many cancers. The VDAC1-based peptides in cells in culture selectively affect cancerous, but not non-cancerous cells, inducing cell death in a variety of cancers, regardless of the cancer origin or genetics. They inhibit cell energy production, eliminate cancer stem cells, and act very rapidly and at low micro-molar concentrations. The activity of these peptides has been validated in several mouse cancer models of glioblastoma, lung, and breast cancers. Their anti-cancer activity involves a multi-pronged attack targeting the hallmarks of cancer. They were also found to be effective in treating non-alcoholic fatty liver disease and diabetes mellitus. Thus, VDAC1-based peptides, by targeting VDAC1-interacting proteins, offer an affordable and innovative new conceptual therapeutic paradigm that can potentially overcome heterogeneity, chemoresistance, and invasive metastatic formation.
- Published
- 2024
- Full Text
- View/download PDF
5. Apoptotic proteins with non-apoptotic activity: expression and function in cancer
- Author
-
Shoshan-Barmatz, Varda, Arif, Tasleem, and Shteinfer-Kuzmine, Anna
- Published
- 2023
- Full Text
- View/download PDF
6. Overexpression of the mitochondrial anti-viral signaling protein, MAVS, in cancers is associated with cell survival and inflammation
- Author
-
Sweta Trishna, Avia Lavon, Anna Shteinfer-Kuzmine, Avis Dafa-Berger, and Varda Shoshan-Barmatz
- Subjects
MT: Non-coding RNAs ,cancer ,inflammation ,MAVS ,mitochondria ,siRNA ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Mitochondrial anti-viral signaling protein (MAVS) plays an important role in host defense against viral infection via coordinating the activation of NF-κB and interferon regulatory factors. The mitochondrial-bound form of MAVS is essential for its anti-viral innate immunity. Recently, tumor cells were proposed to mimic a viral infection by activating RNA-sensing pattern recognition receptors. Here, we demonstrate that MAVS is overexpressed in a panel of viral non-infected cancer cell lines and patient-derived tumors, including lung, liver, bladder, and cervical cancers, and we studied its role in cancer. Silencing MAVS expression reduced cell proliferation and the expression and nuclear translocation of proteins associated with transcriptional regulation, inflammation, and immunity. MAVS depletion reduced expression of the inflammasome components and inhibited its activation/assembly. Moreover, MAVS directly interacts with the mitochondrial protein VDAC1, decreasing its conductance, and we identified the VDAC1 binding site in MAVS. Our findings suggest that MAVS depletion, by reducing cancer cell proliferation and inflammation, represents a new target for cancer therapy.
- Published
- 2023
- Full Text
- View/download PDF
7. Correction: Targeting the overexpressed mitochondrial protein VDAC1 in a mouse model of Alzheimer’s disease protects against mitochondrial dysfunction and mitigates brain pathology
- Author
-
Verma, Ankit, Shteinfer-Kuzmine, Anna, Kamenetsky, Nikita, Pittala, Srinivas, Paul, Avijit, Nahon Crystal, Edna, Ouro, Alberto, Chalifa-Caspi, Vered, Pandey, Swaroop Kumar, Monsonego, Alon, Vardi, Noga, Knafo, Shira, and Shoshan-Barmatz, Varda
- Published
- 2023
- Full Text
- View/download PDF
8. Targeting the overexpressed mitochondrial protein VDAC1 in a mouse model of Alzheimer’s disease protects against mitochondrial dysfunction and mitigates brain pathology
- Author
-
Ankit Verma, Anna Shteinfer-Kuzmine, Nikita Kamenetsky, Srinivas Pittala, Avijit Paul, Edna Nahon Crystal, Alberto Ouro, Vered Chalifa-Caspi, Swaroop Kumar Pandey, Alon Monsengo, Noga Vardi, Shira Knafo, and Varda Shoshan-Barmatz
- Subjects
Alzheimer’s disease ,Metabolism ,Mitochondria ,Neuroinflammation ,VDAC1 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Alzheimer's disease (AD) exhibits mitochondrial dysfunctions associated with dysregulated metabolism, brain inflammation, synaptic loss, and neuronal cell death. As a key protein serving as the mitochondrial gatekeeper, the voltage-dependent anion channel-1 (VDAC1) that controls metabolism and Ca2+ homeostasis is positioned at a convergence point for various cell survival and death signals. Here, we targeted VDAC1 with VBIT-4, a newly developed inhibitor of VDAC1 that prevents its pro-apoptotic activity, and mitochondria dysfunction. Methods To address the multiple pathways involved in AD, neuronal cultures and a 5 × FAD mouse model of AD were treated with VBIT-4. We addressed multiple topics related to the disease and its molecular mechanisms using immunoblotting, immunofluorescence, q-RT-PCR, 3-D structural analysis and several behavioral tests. Results In neuronal cultures, amyloid-beta (Aβ)-induced VDAC1 and p53 overexpression and apoptotic cell death were prevented by VBIT-4. Using an AD-like 5 × FAD mouse model, we showed that VDAC1 was overexpressed in neurons surrounding Aβ plaques, but not in astrocytes and microglia, and this was associated with neuronal cell death. VBIT-4 prevented the associated pathophysiological changes including neuronal cell death, neuroinflammation, and neuro-metabolic dysfunctions. VBIT-4 also switched astrocytes and microglia from being pro-inflammatory/neurotoxic to neuroprotective phenotype. Moreover, VBIT-4 prevented cognitive decline in the 5 × FAD mice as evaluated using several behavioral assessments of cognitive function. Interestingly, VBIT-4 protected against AD pathology, with no significant change in phosphorylated Tau and only a slight decrease in Aβ-plaque load. Conclusions The study suggests that mitochondrial dysfunction with its gatekeeper VDAC1 is a promising target for AD therapeutic intervention, and VBIT-4 is a promising drug candidate for AD treatment.
- Published
- 2022
- Full Text
- View/download PDF
9. Silencing the Mitochondrial Gatekeeper VDAC1 as a Potential Treatment for Bladder Cancer
- Author
-
Belal Alhozeel, Swaroop Kumar Pandey, Anna Shteinfer-Kuzmine, Manikandan Santhanam, and Varda Shoshan-Barmatz
- Subjects
bladder cancer ,mitochondria ,si-RNA ,VDAC1 ,Cytology ,QH573-671 - Abstract
The strategy for treating bladder cancer (BC) depends on whether there is muscle invasion or not, with the latter mostly treated with intravesical therapy, such as with bacillus Calmette–Guérin (BCG). However, BCG treatment is unsuccessful in 70% of patients, who are then subjected to radical cystectomy. Although immune-checkpoint inhibitors have been approved as a second-line therapy for a subset of BC patients, these have failed to meet primary endpoints in clinical trials. Thus, it is crucial to find a new treatment. The mitochondrial gatekeeper protein, the voltage-dependent anion channel 1 (VDAC1), mediates metabolic crosstalk between the mitochondria and cytosol and is involved in apoptosis. It is overexpressed in many cancer types, as shown here for BC, pointing to its significance in high-energy-demanding cancer cells. The BC cell lines UM-UC3 and HTB-5 express high VDAC1 levels compared to other cancer cell lines. VDAC1 silencing in these cells using siRNA that recognizes both human and mouse VDAC1 (si-m/hVDAC1-B) reduces cell viability, mitochondria membrane potential, and cellular ATP levels. Here, we used two BC mouse models: subcutaneous UM-UC3 cells and chemically induced BC using the carcinogen N-Butyl-N-(4-hydroxybutyl) nitrosamine (BBN). Subcutaneous UM-UC3-derived tumors treated with si-m/hVDAC1 showed inhibited tumor growth and reprogrammed metabolism, as reflected in the reduced expression of metabolism-related proteins, including Glut1, hexokinase, citrate synthase, complex-IV, and ATP synthase, suggesting reduced metabolic activity. Furthermore, si-m/hVDAC1-B reduced the expression levels of cancer-stem-cell-related proteins (cytokeratin-14, ALDH1a), modifying the tumor microenvironment, including decreased angiogenesis, extracellular matrix, tumor-associated macrophages, and inhibited epithelial–mesenchymal transition. The BBN-induced BC mouse model showed a clear carcinoma, with damaged bladder morphology and muscle-invasive tumors. Treatment with si-m/hVDAC1-B encapsulated in PLGA-PEI nanoparticles that were administered intravesically directly to the bladder showed a decreased tumor area and less bladder morphology destruction and muscle invasion. Overall, the obtained results point to the potential of si-m/hVDAC1-B as a possible therapeutic tool for treating bladder cancer.
- Published
- 2024
- Full Text
- View/download PDF
10. The role of the mitochondrial protein VDAC1 in inflammatory bowel disease: a potential therapeutic target
- Author
-
Verma, Ankit, Pittala, Srinivas, Alhozeel, Belal, Shteinfer-Kuzmine, Anna, Ohana, Ehud, Gupta, Rajeev, Chung, Jay H., and Shoshan-Barmatz, Varda
- Published
- 2022
- Full Text
- View/download PDF
11. SMAC/Diablo controls proliferation of cancer cells by regulating phosphatidylethanolamine synthesis
- Author
-
Swaroop Kumar Pandey, Avijit Paul, Anna Shteinfer‐Kuzmine, Ran Zalk, Uwe Bunz, and Varda Shoshan‐Barmatz
- Subjects
cancer ,mitochondria ,phosphatidylethanolamine synthesis ,phospholipids ,PSD ,SMAC/Diablo ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
SMAC/Diablo, a pro‐apoptotic protein, yet it is overexpressed in several cancer types. We have described a noncanonical function for SMAC/Diablo as a regulator of lipid synthesis during cancer cell proliferation and development. Here, we explore the molecular mechanism through which SMAC/Diablo regulates phospholipid synthesis. We showed that SMAC/Diablo directly interacts with mitochondrial phosphatidylserine decarboxylase (PSD) and inhibits its catalytic activity during synthesis of phosphatidylethanolamine (PE) from phosphatidylserine (PS). Unlike other phospholipids (PLs), PE is synthesized not only in the endoplasmic reticulum but also in mitochondria. As a result, PSD activity and mitochondrial PE levels were increased in the mitochondria of SMAC/Diablo‐deficient cancer cells, with the total amount of cellular PLs and phosphatidylcholine (PC) being lower as compared to SMAC‐expressing cancer cells. Moreover, in the absence of SMAC/Diablo, PSD inhibited cancer cell proliferation by catalysing the overproduction of mitochondrial PE and depleting the cellular levels of PC, PE and PS. Additionally, we demonstrated that both SMAC/Diablo and PSD colocalization in the nucleus resulted in increased levels of nuclear PE, that acts as a signalling molecule in regulating several nuclear activities. By using a peptide array composed of 768‐peptides derived from 11 SMAC‐interacting proteins, we identified six nuclear proteins ARNT, BIRC2, MAML2, NR4A1, BIRC5 and HTRA2 Five of them also interacted with PSD through motifs that are not involved in SMAC binding. Synthetic peptides carrying the PSD‐interacting motifs of these proteins could bind purified PSD and inhibit the PSD catalytic activity. When targeted specifically to the mitochondria or the nucleus, these synthetic peptides inhibited cancer cell proliferation. To our knowledge, these are the first reported inhibitors of PSD acting also as inhibitors of cancer cell proliferation. Altogether, we demonstrated that phospholipid metabolism and PE synthesis regulated by the SMAC‐PSD interaction are essential for cancer cell proliferation and may be potentially targeted for treating cancer.
- Published
- 2021
- Full Text
- View/download PDF
12. Non-apoptotic activity of the mitochondrial protein SMAC/Diablo in lung cancer: Novel target to disrupt survival, inflammation, and immunosuppression
- Author
-
Swaroop Kumar Pandey, Anna Shteinfer-Kuzmine, Vered Chalifa-Caspi, and Varda Shoshan-Barmatz
- Subjects
SMAC ,cell proliferation ,inflammation ,lung cancer ,immunosuppression ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Mitochondrial SMAC/Diablo induces apoptosis by binding the inhibitor of apoptosis proteins (IAPs), thereby activating caspases and, subsequently, apoptosis. Previously, we found that despite its pro-apoptotic activity, SMAC/Diablo is overexpressed in cancer, and demonstrated that in cancer it possesses new essential and non-apoptotic functions that are associated with regulating phospholipid synthesis including modulating mitochondrial phosphatidylserine decarboxylase activity. Here, we demonstrate additional functions for SMAC/Diablo associated with inflammation and immunity. CRISPR/Cas9 SMAC/Diablo-depleted A549 lung cancer cells displayed inhibited cell proliferation and migration. Proteomics analysis of these cells revealed altered expression of proteins associated with lipids synthesis and signaling, vesicular transport and trafficking, metabolism, epigenetics, the extracellular matrix, cell signaling, and neutrophil-mediated immunity. SMAC-KO A549 cell-showed inhibited tumor growth and proliferation and activated apoptosis. The small SMAC-depleted “tumor” showed a morphology of alveoli-like structures, reversed epithelial-mesenchymal transition, and altered tumor microenvironment. The SMAC-lacking tumor showed reduced expression of inflammation-related proteins such as NF-kB and TNF-α, and of the PD-L1, associated with immune system suppression. These results suggest that SMAC is involved in multiple processes that are essential for tumor growth and progression. Thus, targeting SMAC’s non-canonical function is a potential strategy to treat cancer.
- Published
- 2022
- Full Text
- View/download PDF
13. VDAC1-Based Peptides as Potential Modulators of VDAC1 Interactions with Its Partners and as a Therapeutic for Cancer, NASH, and Diabetes.
- Author
-
Shteinfer-Kuzmine, Anna, Santhanam, Manikandan, and Shoshan-Barmatz, Varda
- Subjects
NON-alcoholic fatty liver disease ,BCL-2 proteins ,MITOCHONDRIAL proteins ,CELL-penetrating peptides ,PEPTIDES - Abstract
This review presents current knowledge related to the voltage-dependent anion channel-1 (VDAC1) as a multi-functional mitochondrial protein that acts in regulating both cell life and death. The location of VDAC1 at the outer mitochondrial membrane (OMM) allows control of metabolic cross-talk between the mitochondria and the rest of the cell, and also enables its interaction with proteins that are involved in metabolic, cell death, and survival pathways. VDAC1′s interactions with over 150 proteins can mediate and regulate the integration of mitochondrial functions with cellular activities. To target these protein–protein interactions, VDAC1-derived peptides have been developed. This review focuses specifically on cell-penetrating VDAC1-based peptides that were developed and used as a "decoy" to compete with VDAC1 for its VDAC1-interacting proteins. These peptides interfere with VDAC1 interactions, for example, with metabolism-associated proteins such as hexokinase (HK), or with anti-apoptotic proteins such as Bcl-2 and Bcl-xL. These and other VDAC1-interacting proteins are highly expressed in many cancers. The VDAC1-based peptides in cells in culture selectively affect cancerous, but not non-cancerous cells, inducing cell death in a variety of cancers, regardless of the cancer origin or genetics. They inhibit cell energy production, eliminate cancer stem cells, and act very rapidly and at low micro-molar concentrations. The activity of these peptides has been validated in several mouse cancer models of glioblastoma, lung, and breast cancers. Their anti-cancer activity involves a multi-pronged attack targeting the hallmarks of cancer. They were also found to be effective in treating non-alcoholic fatty liver disease and diabetes mellitus. Thus, VDAC1-based peptides, by targeting VDAC1-interacting proteins, offer an affordable and innovative new conceptual therapeutic paradigm that can potentially overcome heterogeneity, chemoresistance, and invasive metastatic formation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Mitochondrial VDAC1 Silencing in Urethane-Induced Lung Cancer Inhibits Tumor Growth and Alters Cancer Oncogenic Properties.
- Author
-
Melnikov, Nataly, Pittala, Srinivas, Shteinfer-Kuzmine, Anna, and Shoshan-Barmatz, Varda
- Subjects
MITOCHONDRIA ,RESEARCH funding ,ANTINEOPLASTIC agents ,APOPTOSIS ,MAGNETIC resonance imaging ,FLUORESCENT antibody technique ,TUMOR markers ,XENOGRAFTS ,METABOLIC reprogramming ,GENE expression ,MICE ,INTRAVENOUS therapy ,CELL culture ,LUNG tumors ,DRUG efficacy ,ANIMAL experimentation ,CARCINOGENS ,GROWTH factors ,STEM cells ,CELL size ,STAINS & staining (Microscopy) ,MEMBRANE proteins ,NANOPARTICLES - Abstract
Simple Summary: Cancer cells exhibit several key characteristics, including uncontrolled growth, altered metabolism, enhanced survival mechanisms, and resistance to apoptosis, all of which are crucial for their persistence. In our study, we explored the impact of disrupting the production of mitochondrial gatekeeper protein VDAC1 on lung cancer. We induced lung cancer in mice using the chemical urethane, which closely mimics human lung cancer in terms of genetic mutations and molecular changes. Using MRI to monitor the lung tumors, we found that inhibiting VDAC1 expression in this mouse model led to significant changes: reprogramming of cancer cell metabolism, reduced tumor growth, alterations in the tumor microenvironment, and elimination of cancer stem cells (CSCs). Additionally, treatment with a peptide derived from VDAC1 also inhibited tumor growth and decreased CSC markers. These findings suggest that targeting VDAC1, either through depletion or with a cell-penetrating peptide, could be a promising therapeutic approach for lung cancer. Alterations in cellular metabolism are vital for cancer cell growth and motility. Here, we focused on metabolic reprogramming and changes in tumor hallmarks in lung cancer by silencing the expression of the mitochondrial gatekeeper VDAC1. To better mimic the clinical situation of lung cancer, we induced lung cancer in A/J mice using the carcinogen urethane and examined the effectiveness of si-m/hVDAC1-B encapsulated in PLGA-PEI nanoparticles. si-m/hVDAC1-B, given intravenously, induced metabolism reprogramming and inhibited tumor growth as monitored using MRI. Mice treated with non-targeted (NT) PLGA-PEI-si-NT showed many large size tumors in the lungs, while in PLGA-PEI-si-m/hVDAC-B-treated mice, lung tumor number and area were markedly decreased. Immunofluorescence staining showed decreased expression of VDAC1 and metabolism-related proteins and altered expression of cancer stem cell markers. Morphological analysis showed two types of tumors differing in their morphology; cell size and organization within the tumor. Based on specific markers, the two tumor types were identified as small cell (SCLC) and non-small cell (NSCLC) lung cancer. These two types of tumors were found only in control tumors, suggesting that PLGA-PEI-si-m/hVDAC1-B also targeted SCLC. Indeed, using a xenograft mouse model of human-derived SCLC H69 cells, si-m/hVDAC1-B inhibited tumor growth and reduced the expression of VDAC1 and energy- and metabolism-related enzymes, and of cancer stem cells in the established xenograft. Additionally, intravenous treatment of urethane-induced lung cancer mice with the VDAC1-based peptide, Retro-Tf-D-LP4, showed inhibition of tumor growth, and decreased expression levels of metabolism- and cancer stem cells-related proteins. Thus, silencing VDAC1 targeting both NSCLC and SCLC points to si-VDAC1 as a possible therapeutic tool to treat these lung cancer types. This is important as target NSCLC tumors undergo transformation to SCLC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. The role of the mitochondrial protein VDAC1 in inflammatory bowel disease: a potential therapeutic target
- Author
-
Verma, Ankit, primary, Pittala, Srinivas, additional, Alhozeel, Belal, additional, Shteinfer-Kuzmine, Anna, additional, Ohana, Ehud, additional, Gupta, Rajeev, additional, Chung, Jay H., additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2024
- Full Text
- View/download PDF
16. VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease
- Author
-
Kim, Jeonghan, Gupta, Rajeev, Blanco, Luz P., Yang, Shutong, Shteinfer-Kuzmine, Anna, Wang, Kening, Zhu, Jun, Yoon, Hee Eun, Wang, Xinghao, Kerkhofs, Martijn, Kang, Hyeog, Brown, Alexandra L., Park, Sung-Jun, Xu, Xihui, van Rilland, Eddy Zandee, Kim, Myung K., Cohen, Jeffrey I., Kaplan, Mariana J., Shoshan-Barmatz, Varda, and Chung, Jay H.
- Published
- 2019
17. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target?
- Author
-
Varda Shoshan-Barmatz, Uttpal Anand, Edna Nahon-Crystal, Marta Di Carlo, and Anna Shteinfer-Kuzmine
- Subjects
apoptosis ,cancer ,metabolism ,metformin ,hexokinase ,COVID-19 ,Physiology ,QP1-981 - Abstract
Metformin has been used for treating diabetes mellitus since the late 1950s. In addition to its antihyperglycemic activity, it was shown to be a potential drug candidate for treating a range of other diseases that include various cancers, cardiovascular diseases, diabetic kidney disease, neurodegenerative diseases, renal diseases, obesity, inflammation, COVID-19 in diabetic patients, and aging. In this review, we focus on the important aspects of mitochondrial dysfunction in energy metabolism and cell death with their gatekeeper VDAC1 (voltage-dependent anion channel 1) as a possible metformin target, and summarize metformin’s effects in several diseases and gut microbiota. We question how the same drug can act on diseases with opposite characteristics, such as increasing apoptotic cell death in cancer, while inhibiting it in neurodegenerative diseases. Interestingly, metformin’s adverse effects in many diseases all show VDAC1 involvement, suggesting that it is a common factor in metformin-affecting diseases. The findings that metformin has an opposite effect on various diseases are consistent with the fact that VDAC1 controls cell life and death, supporting the idea that it is a target for metformin.
- Published
- 2021
- Full Text
- View/download PDF
18. The Multicellular Effects of VDAC1 N-Terminal-Derived Peptide
- Author
-
Uttpal Anand, Anna Shteinfer-Kuzmine, Gal Sela, Manikandan Santhanam, Benjamin Gottschalk, Rajaa Boujemaa-Paterski, Ohad Medalia, Wolfgang F. Graier, and Varda Shoshan-Barmatz
- Subjects
apoptosis ,mitochondria ,peptide array ,protein–protein interaction ,voltage-dependent anion channel-1 ,Microbiology ,QR1-502 - Abstract
The mitochondrial voltage-dependent anion channel-1 (VDAC1) protein functions in a variety of mitochondria-linked physiological and pathological processes, including metabolism and cell signaling, as well as in mitochondria-mediated apoptosis. VDAC1 interacts with about 150 proteins to regulate the integration of mitochondrial functions with other cellular activities. Recently, we developed VDAC1-based peptides that have multiple effects on cancer cells and tumors including apoptosis induction. Here, we designed several cell-penetrating VDAC1 N-terminal-derived peptides with the goal of identifying the shortest peptide with improved cellular stability and activity. We identified the D-Δ(1-18)N-Ter-Antp comprising the VDAC1 N-terminal region (19–26 amino acids) fused to the Antp, a cell-penetrating peptide. We demonstrated that this peptide induced apoptosis, autophagy, senescence, cell volume enlargement, and the refusion of divided daughter cells into a single cell, it was responsible for reorganization of actin and tubulin filaments, and increased cell adhesion. In addition, the peptide induced alterations in the expression of proteins associated with cell metabolism, signaling, and division, such as enhancing the expression of nuclear factor kappa B and decreasing the expression of the nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha. These cellular effects may result from the peptide interfering with VDAC1 interaction with its interacting proteins, thereby blocking multiple mitochondrial/VDAC1 pathways associated with cell functions. The results of this study further support the role of VDAC1 as a mitochondrial gatekeeper protein in controlling a variety of cell functions via interaction with associated proteins.
- Published
- 2022
- Full Text
- View/download PDF
19. VDAC1, mitochondrial dysfunction, and Alzheimer's disease
- Author
-
Shoshan-Barmatz, Varda, Nahon-Crystal, Edna, Shteinfer-Kuzmine, Anna, and Gupta, Rajeev
- Published
- 2018
- Full Text
- View/download PDF
20. Silencing VDAC1 to Treat Mesothelioma Cancer: Tumor Reprograming and Altering Tumor Hallmarks
- Author
-
Swaroop Kumar Pandey, Renen Machlof-Cohen, Manikandan Santhanam, Anna Shteinfer-Kuzmine, and Varda Shoshan-Barmatz
- Subjects
mesothelioma ,metabolism ,mitochondria ,VDAC1 ,Microbiology ,QR1-502 - Abstract
Mesothelioma, an aggressive cancer with a poor prognosis, is linked to asbestos exposure. However, carbon nanotubes found in materials we are exposed to daily can cause mesothelioma cancer. Cancer cells reprogram their metabolism to support increased biosynthetic and energy demands required for their growth and motility. Here, we examined the effects of silencing the expression of the voltage-dependent anion channel 1 (VDAC1), controlling the metabolic and energetic crosstalk between mitochondria and the rest of the cell. We demonstrate that VDAC1 is overexpressed in mesothelioma patients; its levels increase with disease stage and are associated with low survival rates. Silencing VDAC1 expression using a specific siRNA identifying both mouse and human VDAC1 (si-m/hVDAC1-B) inhibits cell proliferation of mesothelioma cancer cells. Treatment of xenografts of human-derived H226 cells or mouse-derived AB1 cells with si-m/hVDAC1-B inhibited tumor growth and caused metabolism reprogramming, as reflected in the decreased expression of metabolism-related proteins, including glycolytic and tricarboxylic acid (-)cycle enzymes and the ATP-synthesizing enzyme. In addition, tumors depleted of VDAC1 showed altered microenvironments and inflammation, both associated with cancer progression. Finally, tumor VDAC1 silencing also eliminated cancer stem cells and induced cell differentiation to normal-like cells. The results show that silencing VDAC1 expression leads to reprogrammed metabolism and to multiple effects from tumor growth inhibition to modulation of the tumor microenvironment and inflammation, inducing differentiation of malignant cells. Thus, silencing VDAC1 is a potential therapeutic approach to treating mesothelioma.
- Published
- 2022
- Full Text
- View/download PDF
21. VDAC1 functions in Ca2+ homeostasis and cell life and death in health and disease
- Author
-
Shoshan-Barmatz, Varda, Krelin, Yakov, and Shteinfer-Kuzmine, Anna
- Published
- 2018
- Full Text
- View/download PDF
22. Selective induction of cancer cell death by VDAC1‐based peptides and their potential use in cancer therapy
- Author
-
Anna Shteinfer‐Kuzmine, Zohar Amsalem, Tasleem Arif, Alexandra Zooravlov, and Varda Shoshan‐Barmatz
- Subjects
apoptosis ,cancer ,metabolism ,mitochondria ,peptides ,VDAC1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Mitochondrial VDAC1 mediates cross talk between the mitochondria and other parts of the cell by transporting anions, cations, ATP, Ca2+, and metabolites and serves as a key player in apoptosis. As such, VDAC1 is involved in two important hallmarks of cancer development, namely energy and metabolic reprograming and apoptotic cell death evasion. We previously developed cell‐penetrating VDAC1‐derived peptides that interact with hexokinase (HK), Bcl‐2, and Bcl‐xL to prevent the anti‐apoptotic activities of these proteins and induce cancer cell death, with a focus on leukemia and glioblastoma. In this study, we demonstrated the sensitivity of a panel of genetically characterized cancer cell lines, differing in origin and carried mutations, to VDAC1‐based peptide‐induced apoptosis. Noncancerous cell lines were less affected by the peptides. Furthermore, we constructed additional VDAC1‐based peptides with the aim of improving targeting, selectivity, and cellular stability, including R‐Tf‐D‐LP4, containing the transferrin receptor internalization sequence (Tf) that allows targeting of the peptide to cancer cells, known to overexpress the transferrin receptor. The mode of action of the VDAC1‐based peptides involves HK detachment, interfering with the action of anti‐apoptotic proteins, and thus activating multiple routes leading to an impairment of cell energy and metabolism homeostasis and the induction of apoptosis. Finally, in xenograft glioblastoma, lung, and breast cancer mouse models, R‐Tf‐D‐LP4 inhibited tumor growth while inducing massive cancer cell death, including of cancer stem cells. Thus, VDAC1‐based peptides offer an innovative new conceptual framework for cancer therapy.
- Published
- 2018
- Full Text
- View/download PDF
23. Overexpression of the mitochondrial anti-viral signaling protein, MAVS, in cancers is associated with cell survival and inflammation
- Author
-
Trishna, Sweta, primary, Lavon, Avia, additional, Shteinfer-Kuzmine, Anna, additional, Dafa-Berger, Avis, additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2023
- Full Text
- View/download PDF
24. Silencing the Mitochondrial Gatekeeper VDAC1 as a Potential Treatment for Bladder Cancer.
- Author
-
Alhozeel, Belal, Pandey, Swaroop Kumar, Shteinfer-Kuzmine, Anna, Santhanam, Manikandan, and Shoshan-Barmatz, Varda
- Subjects
BLADDER cancer ,BCG immunotherapy ,EXTRACELLULAR matrix ,ADENOSINE triphosphatase ,CITRATE synthase ,CELL survival ,PLANT mitochondria ,BREAST - Abstract
The strategy for treating bladder cancer (BC) depends on whether there is muscle invasion or not, with the latter mostly treated with intravesical therapy, such as with bacillus Calmette–Guérin (BCG). However, BCG treatment is unsuccessful in 70% of patients, who are then subjected to radical cystectomy. Although immune-checkpoint inhibitors have been approved as a second-line therapy for a subset of BC patients, these have failed to meet primary endpoints in clinical trials. Thus, it is crucial to find a new treatment. The mitochondrial gatekeeper protein, the voltage-dependent anion channel 1 (VDAC1), mediates metabolic crosstalk between the mitochondria and cytosol and is involved in apoptosis. It is overexpressed in many cancer types, as shown here for BC, pointing to its significance in high-energy-demanding cancer cells. The BC cell lines UM-UC3 and HTB-5 express high VDAC1 levels compared to other cancer cell lines. VDAC1 silencing in these cells using siRNA that recognizes both human and mouse VDAC1 (si-m/hVDAC1-B) reduces cell viability, mitochondria membrane potential, and cellular ATP levels. Here, we used two BC mouse models: subcutaneous UM-UC3 cells and chemically induced BC using the carcinogen N-Butyl-N-(4-hydroxybutyl) nitrosamine (BBN). Subcutaneous UM-UC3-derived tumors treated with si-m/hVDAC1 showed inhibited tumor growth and reprogrammed metabolism, as reflected in the reduced expression of metabolism-related proteins, including Glut1, hexokinase, citrate synthase, complex-IV, and ATP synthase, suggesting reduced metabolic activity. Furthermore, si-m/hVDAC1-B reduced the expression levels of cancer-stem-cell-related proteins (cytokeratin-14, ALDH1a), modifying the tumor microenvironment, including decreased angiogenesis, extracellular matrix, tumor-associated macrophages, and inhibited epithelial–mesenchymal transition. The BBN-induced BC mouse model showed a clear carcinoma, with damaged bladder morphology and muscle-invasive tumors. Treatment with si-m/hVDAC1-B encapsulated in PLGA-PEI nanoparticles that were administered intravesically directly to the bladder showed a decreased tumor area and less bladder morphology destruction and muscle invasion. Overall, the obtained results point to the potential of si-m/hVDAC1-B as a possible therapeutic tool for treating bladder cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. A VDAC1-Derived N-Terminal Peptide Inhibits Mutant SOD1-VDAC1 Interactions and Toxicity in the SOD1 Model of ALS
- Author
-
Anna Shteinfer-Kuzmine, Shirel Argueti, Rajeev Gupta, Neta Shvil, Salah Abu-Hamad, Yael Gropper, Jan Hoeber, Andrea Magrì, Angela Messina, Elena N. Kozlova, Varda Shoshan-Barmatz, and Adrian Israelson
- Subjects
ALS ,misfolded SOD1 ,mutant SOD1 ,N-terminal peptide ,VDAC1 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Mutations in superoxide dismutase (SOD1) are the second most common cause of familial amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease caused by the death of motor neurons in the brain and spinal cord. SOD1 neurotoxicity has been attributed to aberrant accumulation of misfolded SOD1, which in its soluble form binds to intracellular organelles, such as mitochondria and ER, disrupting their functions. Here, we demonstrate that mutant SOD1 binds specifically to the N-terminal domain of the voltage-dependent anion channel (VDAC1), an outer mitochondrial membrane protein controlling cell energy, metabolic and survival pathways. Mutant SOD1G93A and SOD1G85R, but not wild type SOD1, directly interact with VDAC1 and reduce its channel conductance. No such interaction with N-terminal-truncated VDAC1 occurs. Moreover, a VDAC1-derived N-terminal peptide inhibited mutant SOD1-induced toxicity. Incubation of motor neuron-like NSC-34 cells expressing mutant SOD1 or mouse embryonic stem cell-derived motor neurons with different VDAC1 N-terminal peptides resulted in enhanced cell survival. Taken together, our results establish a direct link between mutant SOD1 toxicity and the VDAC1 N-terminal domain and suggest that VDAC1 N-terminal peptides targeting mutant SOD1 provide potential new therapeutic strategies for ALS.
- Published
- 2019
- Full Text
- View/download PDF
26. Novel Compounds Targeting the Mitochondrial Protein VDAC1 Inhibit Apoptosis and Protect against Mitochondrial Dysfunction
- Author
-
Ben-Hail, Danya, Begas-Shvartz, Racheli, Shalev, Moran, Shteinfer-Kuzmine, Anna, Gruzman, Arie, Reina, Simona, De Pinto, Vito, and Shoshan-Barmatz, Varda
- Published
- 2016
- Full Text
- View/download PDF
27. Pro-Apoptotic and Anti-Cancer Activity of the Vernonanthura Nudiflora Hydroethanolic Extract
- Author
-
Almog Nadir, Anna Shteinfer-Kuzmine, Swaroop Kumar Pandey, Juan Ortas, Daniel Kerekes, and Varda Shoshan-Barmatz
- Subjects
apoptosis ,cancer ,metabolism ,mitochondria ,plant extract ,VDAC1 ,Cancer Research ,Oncology - Abstract
The mitochondrial voltage-dependent anion channel 1 (VDAC1) protein is involved in several essential cancer hallmarks, including energy and metabolism reprogramming and apoptotic cell death evasion. In this study, we demonstrated the ability of hydroethanolic extracts from three different plants, Vernonanthura nudiflora (Vern), Baccharis trimera (Bac), and Plantago major (Pla), to induce cell death. We focused on the most active Vern extract. We demonstrated that it activates multiple pathways that lead to impaired cell energy and metabolism homeostasis, elevated ROS production, increased intracellular Ca2+, and mitochondria-mediated apoptosis. The massive cell death generated by this plant extract’s active compounds involves the induction of VDAC1 overexpression and oligomerization and, thereby, apoptosis. Gas chromatography of the hydroethanolic plant extract identified dozens of compounds, including phytol and ethyl linoleate, with the former producing similar effects as the Vern hydroethanolic extract but at 10-fold higher concentrations than those found in the extract. In a xenograft glioblastoma mouse model, both the Vern extract and phytol strongly inhibited tumor growth and cell proliferation and induced massive tumor cell death, including of cancer stem cells, inhibiting angiogenesis and modulating the tumor microenvironment. Taken together, the multiple effects of Vern extract make it a promising potential cancer therapeutic.
- Published
- 2023
- Full Text
- View/download PDF
28. VDAC1 at the Intersection of Cell Metabolism, Apoptosis, and Diseases
- Author
-
Varda Shoshan-Barmatz, Anna Shteinfer-Kuzmine, and Ankit Verma
- Subjects
apoptosis ,cancer ,diseases ,metabolism ,mitochondria ,VDAC1 ,Microbiology ,QR1-502 - Abstract
The voltage-dependent anion channel 1 (VDAC1) protein, is an important regulator of mitochondrial function, and serves as a mitochondrial gatekeeper, with responsibility for cellular fate. In addition to control over energy sources and metabolism, the protein also regulates epigenomic elements and apoptosis via mediating the release of apoptotic proteins from the mitochondria. Apoptotic and pathological conditions, as well as certain viruses, induce cell death by inducing VDAC1 overexpression leading to oligomerization, and the formation of a large channel within the VDAC1 homo-oligomer. This then permits the release of pro-apoptotic proteins from the mitochondria and subsequent apoptosis. Mitochondrial DNA can also be released through this channel, which triggers type-Ι interferon responses. VDAC1 also participates in endoplasmic reticulum (ER)-mitochondria cross-talk, and in the regulation of autophagy, and inflammation. Its location in the outer mitochondrial membrane, makes VDAC1 ideally placed to interact with over 100 proteins, and to orchestrate the interaction of mitochondrial and cellular activities through a number of signaling pathways. Here, we provide insights into the multiple functions of VDAC1 and describe its involvement in several diseases, which demonstrate the potential of this protein as a druggable target in a wide variety of pathologies, including cancer.
- Published
- 2020
- Full Text
- View/download PDF
29. Targeting the overexpressed mitochondrial protein VDAC1 in a mouse model of Alzheimer’s disease protects against mitochondrial dysfunction and mitigates brain pathology
- Author
-
Verma, Ankit, primary, Shteinfer-Kuzmine, Anna, additional, Kamenetsky, Nikita, additional, Pittala, Srinivas, additional, Paul, Avijit, additional, Nahon Crystal, Edna, additional, Ouro, Alberto, additional, Chalifa-Caspi, Vered, additional, Pandey, Swaroop Kumar, additional, Monsonego, Alon, additional, Vardi, Noga, additional, Knafo, Shira, additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2022
- Full Text
- View/download PDF
30. Voltage-Dependent Anion Channel 1 As an Emerging Drug Target for Novel Anti-Cancer Therapeutics
- Author
-
Varda Shoshan-Barmatz, Yakov Krelin, Anna Shteinfer-Kuzmine, and Tasleem Arif
- Subjects
apoptosis ,cancer ,metabolism ,mitochondria ,voltage-dependent anion channel 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Cancer cells share several properties, high proliferation potential, reprogramed metabolism, and resistance to apoptotic cues. Acquiring these hallmarks involves changes in key oncogenes and non-oncogenes essential for cancer cell survival and prosperity, and is accompanied by the increased energy requirements of proliferating cells. Mitochondria occupy a central position in cell life and death with mitochondrial bioenergetics, biosynthesis, and signaling are critical for tumorigenesis. Voltage-dependent anion channel 1 (VDAC1) is situated in the outer mitochondrial membrane (OMM) and serving as a mitochondrial gatekeeper. VDAC1 allowing the transfer of metabolites, fatty acid ions, Ca2+, reactive oxygen species, and cholesterol across the OMM and is a key player in mitochondrial-mediate apoptosis. Moreover, VDAC1 serves as a hub protein, interacting with diverse sets of proteins from the cytosol, endoplasmic reticulum, and mitochondria that together regulate metabolic and signaling pathways. The observation that VDAC1 is over-expressed in many cancers suggests that the protein may play a pivotal role in cancer cell survival. However, VDAC1 is also important in mitochondria-mediated apoptosis, mediating release of apoptotic proteins and interacting with anti-apoptotic proteins, such as B-cell lymphoma 2 (Bcl-2), Bcl-xL, and hexokinase (HK), which are also highly expressed in many cancers. Strategically located in a “bottleneck” position, controlling metabolic homeostasis and apoptosis, VDAC1 thus represents an emerging target for anti-cancer drugs. This review presents an overview on the multi-functional mitochondrial protein VDAC1 performing several functions and interacting with distinct sets of partners to regulate both cell life and death, and highlights the importance of the protein for cancer cell survival. We address recent results related to the mechanisms of VDAC1-mediated apoptosis and the potential of associated proteins to modulate of VDAC1 activity, with the aim of developing VDAC1-based approaches. The first strategy involves modification of cell metabolism using VDAC1-specific small interfering RNA leading to inhibition of cancer cell and tumor growth and reversed oncogenic properties. The second strategy involves activation of cancer cell death using VDAC1-based peptides that prevent cell death induction by anti-apoptotic proteins. Finally, we discuss the potential therapeutic benefits of treatments and drugs leading to enhanced VDAC1 expression or targeting VDAC1 to induce apoptosis.
- Published
- 2017
- Full Text
- View/download PDF
31. The Multicellular Effects of VDAC1 N-Terminal-Derived Peptide
- Author
-
Anand, Uttpal, primary, Shteinfer-Kuzmine, Anna, additional, Sela, Gal, additional, Santhanam, Manikandan, additional, Gottschalk, Benjamin, additional, Boujemaa-Paterski, Rajaa, additional, Medalia, Ohad, additional, Graier, Wolfgang F., additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2022
- Full Text
- View/download PDF
32. Non-apoptotic activity of the mitochondrial protein SMAC/Diablo in lung cancer: Novel target to disrupt survival, inflammation, and immunosuppression
- Author
-
Pandey, Swaroop Kumar, primary, Shteinfer-Kuzmine, Anna, additional, Chalifa-Caspi, Vered, additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2022
- Full Text
- View/download PDF
33. Targeting the Mitochondrial Protein VDAC1 as a Potential Therapeutic Strategy in ALS
- Author
-
Shteinfer-Kuzmine, Anna, primary, Argueti-Ostrovsky, Shirel, additional, Leyton-Jaimes, Marcel F., additional, Anand, Uttpal, additional, Abu-Hamad, Salah, additional, Zalk, Ran, additional, Shoshan-Barmatz, Varda, additional, and Israelson, Adrian, additional
- Published
- 2022
- Full Text
- View/download PDF
34. Silencing VDAC1 to Treat Mesothelioma Cancer: Tumor Reprograming and Altering Tumor Hallmarks
- Author
-
Pandey, Swaroop Kumar, primary, Machlof-Cohen, Renen, additional, Santhanam, Manikandan, additional, Shteinfer-Kuzmine, Anna, additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2022
- Full Text
- View/download PDF
35. Pro-Apoptotic and Anti-Cancer Activity of the Vernonanthura Nudiflora Hydroethanolic Extract.
- Author
-
Nadir, Almog, Shteinfer-Kuzmine, Anna, Pandey, Swaroop Kumar, Ortas, Juan, Kerekes, Daniel, and Shoshan-Barmatz, Varda
- Subjects
MEDICINAL plants ,XENOGRAFTS ,ELECTROPHORESIS ,APOPTOSIS ,WATER ,PLANTS ,MITOCHONDRIA ,CELL survival ,IMMUNOBLOTTING ,GAS chromatography ,AGAR ,MASS spectrometry ,FLUORESCENT antibody technique ,CELL proliferation ,STEM cells ,RESEARCH funding ,PLANT extracts ,TUMORS ,CELL lines ,REACTIVE oxygen species ,MEMBRANE proteins ,ETHANOL ,CARRIER proteins - Abstract
Simple Summary: Natural products derived from plants have numerous clinical applications, including anti-cancer activity. In the present study, we identified three different plant extracts as strong inducers of cell death that were not reported previously. We focused on the most potent of these plants, Vernonanthura nudiflora (Vern). We demonstrated that the plant extracts obtained by treatment with a water and ethanol mixture killed tumor cells via multiple routes. These include impairing cell energy and metabolism, generating reactive oxygen species, increasing intracellular Ca
2+ , and inducing mitochondria-mediated apoptosis. We connected these activities to increased levels of the mitochondrial gatekeeper protein, VDAC1, which is associated with metabolism and apoptosis regulation. In a glioblastoma mouse model, Vern extract strongly inhibited tumor growth and induced massive tumor cell death, including cancer stem cells, by inhibiting blood supply and modulating the tumor microenvironment. The multipronged effects of hydroethanolic Vern extract make it a promising candidate for treating cancer. The mitochondrial voltage-dependent anion channel 1 (VDAC1) protein is involved in several essential cancer hallmarks, including energy and metabolism reprogramming and apoptotic cell death evasion. In this study, we demonstrated the ability of hydroethanolic extracts from three different plants, Vernonanthura nudiflora (Vern), Baccharis trimera (Bac), and Plantago major (Pla), to induce cell death. We focused on the most active Vern extract. We demonstrated that it activates multiple pathways that lead to impaired cell energy and metabolism homeostasis, elevated ROS production, increased intracellular Ca2+ , and mitochondria-mediated apoptosis. The massive cell death generated by this plant extract's active compounds involves the induction of VDAC1 overexpression and oligomerization and, thereby, apoptosis. Gas chromatography of the hydroethanolic plant extract identified dozens of compounds, including phytol and ethyl linoleate, with the former producing similar effects as the Vern hydroethanolic extract but at 10-fold higher concentrations than those found in the extract. In a xenograft glioblastoma mouse model, both the Vern extract and phytol strongly inhibited tumor growth and cell proliferation and induced massive tumor cell death, including of cancer stem cells, inhibiting angiogenesis and modulating the tumor microenvironment. Taken together, the multiple effects of Vern extract make it a promising potential cancer therapeutic. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
36. Targeting the Mitochondrial Protein VDAC1 as a Potential Therapeutic Strategy in ALS
- Author
-
Anna Shteinfer-Kuzmine, Shirel Argueti-Ostrovsky, Marcel F. Leyton-Jaimes, Uttpal Anand, Salah Abu-Hamad, Ran Zalk, Varda Shoshan-Barmatz, and Adrian Israelson
- Subjects
Superoxide Dismutase ,VDAC1 ,ALS ,mutant SOD1 ,mitochondria ,apoptosis ,misfolded proteins ,Voltage-Dependent Anion Channel 1 ,Amyotrophic Lateral Sclerosis ,Organic Chemistry ,Mice, Transgenic ,General Medicine ,Catalysis ,Rats ,Computer Science Applications ,Mitochondrial Proteins ,Inorganic Chemistry ,Disease Models, Animal ,Mice ,Superoxide Dismutase-1 ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy - Abstract
Impaired mitochondrial function has been proposed as a causative factor in neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), caused by motor neuron degeneration. Mutations in superoxide dismutase (SOD1) cause ALS and SOD1 mutants were shown to interact with the voltage-dependent anion channel 1 (VDAC1), affecting its normal function. VDAC1 is a multi-functional channel located at the outer mitochondrial membrane that serves as a mitochondrial gatekeeper controlling metabolic and energetic crosstalk between mitochondria and the rest of the cell and it is a key player in mitochondria-mediated apoptosis. Previously, we showed that VDAC1 interacts with SOD1 and that the VDAC1-N-terminal-derived peptide prevented mutant SOD1 cytotoxic effects. In this study, using a peptide array, we identified the SOD1 sequence that interacts with VDAC1. Synthetic peptides generated from the identified VDAC1-binding sequences in SOD1 directly interacted with purified VDAC1. We also show that VDAC1 oligomerization increased in spinal cord mitochondria isolated from mutant SOD1G93A mice and rats. Thus, we used the novel VDAC1-specific small molecules, VBIT-4 and VBIT-12, inhibiting VDAC1 oligomerization and subsequently apoptosis and associated processes such as ROS production, and increased cytosolic Ca2+. VBIT-12 was able to rescue cell death induced by mutant SOD1 in neuronal cultures. Finally, although survival was not affected, VBIT-12 administration significantly improved muscle endurance in mutant SOD1G93A mice. Therefore, VBIT-12 may represent an attractive therapy for maintaining muscle function during the progression of ALS.
- Published
- 2022
- Full Text
- View/download PDF
37. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target?
- Author
-
Shoshan-Barmatz, Varda, primary, Anand, Uttpal, additional, Nahon-Crystal, Edna, additional, Di Carlo, Marta, additional, and Shteinfer-Kuzmine, Anna, additional
- Published
- 2021
- Full Text
- View/download PDF
38. VDAC1 Silencing in Cancer Cells Leads to Metabolic Reprogramming That Modulates Tumor Microenvironment
- Author
-
Vered Chalifa-Caspi, Erez Zerbib, Anna Shteinfer-Kuzmine, Tasleem Arif, and Varda Shoshan-Barmatz
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Angiogenesis ,Biology ,Article ,Malignant transformation ,03 medical and health sciences ,0302 clinical medicine ,tumor microenvironment ,RC254-282 ,Tumor microenvironment ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,reprogramming ,Cell biology ,mitochondria ,VDAC1 ,030104 developmental biology ,Oncology ,Tumor progression ,siRNA ,030220 oncology & carcinogenesis ,Cancer cell ,metabolism ,Reprogramming ,Extracellular matrix organization - Abstract
Simple Summary Tumors are comprised of proliferating cancer cells, and their microenvironment consists of the extracellular matrix, blood vessels, and a variety of tissue cells. The tumor microenvironment functions in cell growth, proliferation, migration, immunity, malignant transformation, and apoptosis. Understanding the molecular interactions between cancer cells and their microenvironment would facilitate the development of therapeutic strategies to disrupt these interactions and fight cancer. Here, we demonstrate that depleting the mitochondrial gatekeeper VDAC1 in human cancer cells in tumors led to metabolic reprogramming, inhibited tumor growth, and disrupted tumor–host interactions. A next-generation sequencing analysis of human lung cell-derived tumors expressing or depleted of VDAC1 allows distinguishing genes of human or murine origin, thus enabling the separation of the bidirectional cross-talk between malignant cells and the tumor microenvironment. A battery of human cancer cell and mouse genes associated with tumor microenvironment formation and remodeling were altered. The results point to VDAC1 as a novel target for both inhibiting tumor growth and modulating the tumor microenvironment, thus influencing cancer progression, migration, and invasion. Abstract The tumor microenvironment (TME) plays an important role in cell growth, proliferation, migration, immunity, malignant transformation, and apoptosis. Thus, better insight into tumor–host interactions is required. Most of these processes involve the metabolic reprogramming of cells. Here, we focused on this reprogramming in cancerous cells and its effect on the TME. A major limitation in the study of tumor–host interactions is the difficulty in separating cancerous from non-cancerous signaling pathways within a tumor. Our strategy involved specifically silencing the expression of VDAC1 in the mitochondria of human-derived A549 lung cancer xenografts in mice, but not in the mouse-derived cells of the TME. Next-generation sequencing (NGS) analysis allows distinguishing the human or mouse origin of genes, thus enabling the separation of the bidirectional cross-talk between the TME and malignant cells. We demonstrate that depleting VDAC1 in cancer cells led to metabolic reprogramming, tumor regression, and the disruption of tumor–host interactions. This was reflected in the altered expression of a battery of genes associated with TME, including those involved in extracellular matrix organization and structure, matrix-related peptidases, angiogenesis, intercellular interacting proteins, integrins, and growth factors associated with stromal activities. We show that metabolic rewiring upon mitochondrial VDAC1 silencing in cancer cells affected several components of the TME, such as structural protein matrix metalloproteinases and Lox, and elicited a stromal response resembling the reaction to a foreign body in wound healing. As tumor progression requires a cooperative interplay between the host and cancer cells, and the ECM is intensively remodeled during cancer progression, VDAC1 depletion induced metabolic reprogramming that targeted both tumor cells and resulted in the alteration of the whole spectrum of TME-related genes, affecting the reciprocal feedback between ECM molecules, host cells, and cancer cells. Thus, VDAC1 depletion using si-VDAC1 represents therapeutic potential, inhibiting cancer cell proliferation and also inducing the modulation of TME components, which influences cancer progression, migration, and invasion.
- Published
- 2021
39. Author response for 'SMAC/Diablo controls proliferation of cancer cells by regulating phosphatidylethanolamine synthesis'
- Author
-
Swaroop Kumar Pandey, Uwe H. F. Bunz, Anna Shteinfer-Kuzmine, Avijit Paul, Ran Zalk, and Varda Shoshan‐Barmat
- Subjects
Chemistry ,Cancer cell ,Cancer research ,Smac diablo ,Phosphatidylethanolamine synthesis - Published
- 2021
- Full Text
- View/download PDF
40. VDAC1 Silencing in Cancer Cells Leads to Metabolic Reprogramming That Modulates Tumor Microenvironment
- Author
-
Zerbib, Erez, primary, Arif, Tasleem, additional, Shteinfer-Kuzmine, Anna, additional, Chalifa-Caspi, Vered, additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2021
- Full Text
- View/download PDF
41. SMAC/Diablo controls proliferation of cancer cells by regulating phosphatidylethanolamine synthesis
- Author
-
Pandey, Swaroop Kumar, primary, Paul, Avijit, additional, Shteinfer‐Kuzmine, Anna, additional, Zalk, Ran, additional, Bunz, Uwe, additional, and Shoshan‐Barmatz, Varda, additional
- Published
- 2021
- Full Text
- View/download PDF
42. The role of the mitochondrial protein VDAC1 in inflammatory bowel disease: a potential therapeutic target
- Author
-
Rajeev Gupta, Anna Shteinfer-Kuzmine, Varda Shoshan-Barmatz, Belal Alhozeel, Jay H. Chung, Srinivas Pittala, Ankit Verma, and Ehud Ohana
- Subjects
Inflammasomes ,medicine.medical_treatment ,Mitochondrion ,Inflammatory bowel disease ,Mitochondrial Proteins ,Mice ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Pharmacology ,Chemistry ,Voltage-Dependent Anion Channel 1 ,Dextran Sulfate ,Inflammasome ,medicine.disease ,Colitis ,Inflammatory Bowel Diseases ,Ulcerative colitis ,digestive system diseases ,Epithelium ,Mitochondria ,Cytokine ,medicine.anatomical_structure ,Apoptosis ,Cancer research ,Molecular Medicine ,Original Article ,VDAC1 ,medicine.drug - Abstract
Recent studies have implicated mitochondrial dysfunction as a trigger of inflammatory bowel diseases, including Crohn’s disease (CD) and ulcerative colitis (UC). We have investigated the role of the mitochondria gate-keeper protein, the voltage-dependent-anion channel 1 (VDAC1), in gastrointestinal inflammation and tested the effects of the newly developed VDAC1-interacting molecules, VBIT-4 and VBIT-12, on UC induced by dextran sulfate sodium (DSS) or trinitrobenzene sulphonic acid (TNBS) in mice. VDAC1, which controls metabolism, lipids transport, apoptosis, and inflammasome activation, is overexpressed in the colon of CD and UC patients and DSS-treated mice. VBIT-12 treatment of cultured colon cells inhibited the DSS-induced VDAC1 overexpression, oligomerization, and apoptosis. In the DSS-treated mice, VBIT-12 suppressed weight loss, diarrhea, rectal bleeding, pro-inflammatory cytokine production, crypt and epithelial cell damage, and focal inflammation. VBIT-12 also inhibited the infiltration of inflammatory cells, apoptosis, mtDNA release, and activation of caspase-1 and NRLP3 inflammasome to reduce the inflammatory response. The levels of the ATP-gated P(2)X(7)-Ca(2+)/K(+) channel and ER-IP3R-Ca(2+) channel, and of the mitochondrial anti-viral protein (MAVS), mediating NLRP3 inflammasome assembly and activation, were highly increased in DSS-treated mice, but not when VBIT-12 treated. We conclude that UC may be promoted by VDAC1-overexpression and may therefore be amenable to treatment with novel VDAC1-interacting molecules. This VDAC1-based strategy exploits a completely new target for UC treatment and opens a new avenue for treating other inflammatory/autoimmune diseases.
- Published
- 2020
43. Mitochondria and nucleus cross-talk: Signaling in metabolism, apoptosis, and differentiation, and function in cancer
- Author
-
Anna Shteinfer-Kuzmine, Varda Shoshan-Barmaz, Ankit Verma, Avijit Paul, Tasleem Arif, and Or Aizenberg
- Subjects
0301 basic medicine ,Male ,Clinical Biochemistry ,Mice, Nude ,Apoptosis ,Mitochondrion ,Biochemistry ,Epigenesis, Genetic ,Histones ,Mitochondrial Proteins ,03 medical and health sciences ,0302 clinical medicine ,Receptors, GABA ,Genetics ,medicine ,Animals ,Humans ,Epigenetics ,Molecular Biology ,Cell Nucleus ,biology ,Chemistry ,Brain Neoplasms ,Voltage-Dependent Anion Channel 1 ,Cytochromes c ,Cell Differentiation ,Cell Biology ,Subcellular localization ,Xenograft Model Antitumor Assays ,Cell biology ,030104 developmental biology ,Histone ,medicine.anatomical_structure ,Acetylation ,030220 oncology & carcinogenesis ,Caspases ,biology.protein ,Lymphoma, Large B-Cell, Diffuse ,Signal transduction ,Tumor Suppressor Protein p53 ,Apoptosis Regulatory Proteins ,Glioblastoma ,VDAC1 ,Nucleus - Abstract
The cross-talk between the mitochondrion and the nucleus regulates cellular functions, including differentiation and adaptation to stress. Mitochondria supply metabolites for epigenetic modifications and other nuclear-associated activities and certain mitochondrial proteins were found in the nucleus. The voltage-dependent anion channel 1 (VDAC1), localized at the outer mitochondrial membrane (OMM) is a central protein in controlling energy production, cell growth, Ca2+ homeostasis, and apoptosis. To alter the cross-talk between the mitochondria and the nucleus, we used specific siRNA to silence the expression of VDAC1 in glioblastoma (GBM) U87-MG and U118-MG cell-derived tumors, and then monitored the nuclear localization of mitochondrial proteins and the methylation and acetylation of histones. Depletion of VDAC1 from tumor cells reduced metabolism, leading to inhibition of tumor growth, and several tumor-associated processes and signaling pathways linked to cancer development. In addition, we demonstrate that certain mitochondrial pro-apoptotic proteins such as caspases 3, 8, and 9, and p53 were unexpectedly overexpressed in tumors, suggesting that they possess additional non-apoptotic functions. VDAC1 depletion and metabolic reprograming altered their expression levels and subcellular localization, specifically their translocation to the nucleus. In addition, VDAC1 depletion also leads to epigenetic modifications of histone acetylation and methylation, suggesting that the interchange between metabolism and cancer signaling pathways involves mitochondria-nucleus cross-talk. The mechanisms regulating mitochondrial protein trafficking into and out of the nucleus and the role these proteins play in the nucleus remain to be elucidated.
- Published
- 2020
44. VDAC1 at the Intersection of Cell Metabolism, Apoptosis, and Diseases
- Author
-
Ankit Verma, Varda Shoshan-Barmatz, and Anna Shteinfer-Kuzmine
- Subjects
0301 basic medicine ,Mitochondrial DNA ,Programmed cell death ,lcsh:QR1-502 ,Review ,virus ,Mitochondrion ,Biochemistry ,lcsh:Microbiology ,diseases ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,cancer ,Humans ,Molecular Biology ,Chemistry ,Endoplasmic reticulum ,Voltage-Dependent Anion Channel 1 ,Autophagy ,apoptosis ,Cell biology ,mitochondria ,VDAC1 ,030104 developmental biology ,030220 oncology & carcinogenesis ,Signal transduction ,Energy source ,metabolism - Abstract
The voltage-dependent anion channel 1 (VDAC1) protein, is an important regulator of mitochondrial function, and serves as a mitochondrial gatekeeper, with responsibility for cellular fate. In addition to control over energy sources and metabolism, the protein also regulates epigenomic elements and apoptosis via mediating the release of apoptotic proteins from the mitochondria. Apoptotic and pathological conditions, as well as certain viruses, induce cell death by inducing VDAC1 overexpression leading to oligomerization, and the formation of a large channel within the VDAC1 homo-oligomer. This then permits the release of pro-apoptotic proteins from the mitochondria and subsequent apoptosis. Mitochondrial DNA can also be released through this channel, which triggers type-Ι interferon responses. VDAC1 also participates in endoplasmic reticulum (ER)-mitochondria cross-talk, and in the regulation of autophagy, and inflammation. Its location in the outer mitochondrial membrane, makes VDAC1 ideally placed to interact with over 100 proteins, and to orchestrate the interaction of mitochondrial and cellular activities through a number of signaling pathways. Here, we provide insights into the multiple functions of VDAC1 and describe its involvement in several diseases, which demonstrate the potential of this protein as a druggable target in a wide variety of pathologies, including cancer.
- Published
- 2020
45. The Mitochondrial Protein VDAC1 at the Crossroads of Cancer Cell Metabolism: The Epigenetic Link
- Author
-
Zohar Amsalem, Vered Chalifa-Caspi, Tasleem Arif, Anna Shteinfer-Kuzmine, and Varda Shoshan-Barmatz
- Subjects
0301 basic medicine ,Cancer Research ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,medicine ,cancer ,Epigenetics ,Transcription factor ,histones epigenetics ,Epigenome ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,mitochondria ,VDAC1 ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Signal transduction ,Carcinogenesis ,metabolism - Abstract
Carcinogenesis is a complicated process that involves the deregulation of epigenetics, resulting in cellular transformational events, such as proliferation, differentiation, and metastasis. Most chromatin-modifying enzymes utilize metabolites as co-factors or substrates and thus are directly dependent on such metabolites as acetyl-coenzyme A, S-adenosylmethionine, and NAD+. Here, we show that using specific siRNA to deplete a tumor of VDAC1 not only led to reprograming of the cancer cell metabolism but also altered several epigenetic-related enzymes and factors. VDAC1, in the outer mitochondrial membrane, controls metabolic cross-talk between the mitochondria and the rest of the cell, thus regulating the metabolic and energetic functions of mitochondria, and has been implicated in apoptotic-relevant events. We previously demonstrated that silencing VDAC1 expression in glioblastoma (GBM) U-87MG cell-derived tumors, resulted in reprogramed metabolism leading to inhibited tumor growth, angiogenesis, epithelial&ndash, mesenchymal transition and invasiveness, and elimination of cancer stem cells, while promoting the differentiation of residual tumor cells into neuronal-like cells. These VDAC1 depletion-mediated effects involved alterations in transcription factors regulating signaling pathways associated with cancer hallmarks. As the epigenome is sensitive to cellular metabolism, this study was designed to assess whether depleting VDAC1 affects the metabolism&ndash, epigenetics axis. Using DNA microarrays, q-PCR, and specific antibodies, we analyzed the effects of si-VDAC1 treatment of U-87MG-derived tumors on histone modifications and epigenetic-related enzyme expression levels, as well as the methylation and acetylation state, to uncover any alterations in epigenetic properties. Our results demonstrate that metabolic rewiring of GBM via VDAC1 depletion affects epigenetic modifications, and strongly support the presence of an interplay between metabolism and epigenetics.
- Published
- 2020
46. Mitochondria and nucleus cross‐talk: Signaling in metabolism, apoptosis, and differentiation, and function in cancer
- Author
-
Shteinfer‐Kuzmine, Anna, primary, Verma, Ankit, additional, Arif, Tasleem, additional, Aizenberg, Or, additional, Paul, Avijit, additional, and Shoshan‐Barmaz, Varda, additional
- Published
- 2020
- Full Text
- View/download PDF
47. VDAC1 at the Intersection of Cell Metabolism, Apoptosis, and Diseases
- Author
-
Shoshan-Barmatz, Varda, primary, Shteinfer-Kuzmine, Anna, additional, and Verma, Ankit, additional
- Published
- 2020
- Full Text
- View/download PDF
48. The Mitochondrial Protein VDAC1 at the Crossroads of Cancer Cell Metabolism: The Epigenetic Link
- Author
-
Amsalem, Zohar, primary, Arif, Tasleem, additional, Shteinfer-Kuzmine, Anna, additional, Chalifa-Caspi, Vered, additional, and Shoshan-Barmatz, Varda, additional
- Published
- 2020
- Full Text
- View/download PDF
49. Mitochondrial VDAC1-based peptides: Attacking oncogenic properties in glioblastoma
- Author
-
Varda Shoshan-Barmatz, Shambhoo Sharan Tripathi, Avijit Paul, Anna Shteinfer-Kuzmine, Yakov Krelin, and Tasleem Arif
- Subjects
0301 basic medicine ,Cell Survival ,Cell ,Gene Expression ,Transferrin receptor ,Apoptosis ,Cell-Penetrating Peptides ,Mitochondrion ,Models, Biological ,03 medical and health sciences ,Mice ,Adenosine Triphosphate ,Cell Line, Tumor ,Hexokinase ,medicine ,Animals ,Humans ,Caspase ,chemistry.chemical_classification ,Membrane Potential, Mitochondrial ,biology ,Cytochrome c ,Voltage-Dependent Anion Channel 1 ,glioblastoma ,Cytochromes c ,Mitochondria ,VDAC1 ,Disease Models, Animal ,030104 developmental biology ,Enzyme ,medicine.anatomical_structure ,Oncology ,chemistry ,Immunology ,biology.protein ,Cancer research ,peptides ,Neoplastic Stem Cells ,Apoptosis Regulatory Proteins ,Research Paper - Abstract
// Anna Shteinfer-Kuzmine 1, * , Tasleem Arif 1, * , Yakov Krelin 1 , Shambhoo Sharan Tripathi 1 , Avijit Paul 1 and Varda Shoshan-Barmatz 1 1 Department of Life Sciences and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel * First co-authors Correspondence to: Varda Shoshan-Barmatz, email: vardasb@bgu.ac.il Keywords: apoptosis, glioblastoma, mitochondria, peptides, VDAC1 Received: June 29, 2016 Accepted: November 21, 2016 Published: February 17, 2017 ABSTRACT Glioblastoma multiforme (GBM), a primary brain malignancy characterized by high morbidity, invasiveness, proliferation, relapse and mortality, is resistant to chemo- and radiotherapies and lacks effective treatment. GBM tumors undergo metabolic reprograming and develop anti-apoptotic defenses. We targeted GBM with a peptide derived from the mitochondrial protein voltage-dependent anion channel 1 (VDAC1), a key component of cell energy, metabolism and apoptosis regulation. VDAC1-based cell-penetrating peptides perturbed cell energy and metabolic homeostasis and induced apoptosis in several GBM and GBM-derived stem cell lines. We found that the peptides simultaneously attacked several oncogenic properties of human U-87MG cells introduced into sub-cutaneous xenograft mouse model, inhibiting tumor growth, invasion, and cellular metabolism, stemness and inducing apoptosis. Peptide-treated tumors showed decreased expression of all tested metabolism-related enzymes and transporters, and elevated levels of apoptotic proteins, such as p53, cytochrome c and caspases. Retro-Tf-D-LP4, containing the human transferrin receptor (TfR)-recognition sequence, crossed the blood-brain barrier (BBB) via the TfR that is highly expressed in the BBB to strongly inhibit tumor growth in an intracranial xenograft mouse model. In summary, the VDAC1-based peptides tested here offer a potentially affordable and innovative new conceptual therapeutic paradigm that might overcome GBM stemness and invasiveness and reduce relapse rates.
- Published
- 2017
50. Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States
- Author
-
Tasleem Arif, Anna Shteinfer-Kuzmine, Varda Shoshan-Barmatz, Yakov Krelin, and Avijit Paul
- Subjects
0301 basic medicine ,cancer stem cells ,Cancer Research ,Cellular differentiation ,Cell ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Cancer stem cell ,medicine ,Gene silencing ,PI3K/AKT/mTOR pathway ,si-RNA ,Cancer ,differentiation ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,mitochondria ,VDAC1 ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cancer cell ,Cancer research - Abstract
Oncogenic properties, along with the metabolic reprogramming necessary for tumour growth and motility, are acquired by cancer cells. Thus, tumour metabolism is becoming a target for cancer therapy. Here, cancer cell metabolism was tackled by silencing the expression of voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein that controls cell energy, as well as metabolic and survival pathways and that is often over-expressed in many cancers. We demonstrated that silencing VDAC1 expression using human-specific siRNA (si-hVDAC1) inhibited cancer cell growth, both in vitro and in mouse xenograft models of human glioblastoma (U-87MG), lung cancer (A549), and triple negative breast cancer (MDA-MB-231). Importantly, treatment with si-hVDAC1 induced metabolic rewiring of the cancer cells, reversing their oncogenic properties and diverting them towards differentiated-like cells. The si-hVDAC1-treated residual &ldquo, tumour&rdquo, showed reprogrammed metabolism, decreased proliferation, inhibited stemness and altered expression of genes and proteins, leading to cell differentiation toward less malignant lineages. These VDAC1 depletion-mediated effects involved alterations in master transcription factors associated with cancer hallmarks, such as highly increased expression of p53 and decreased expression of HIF-1a and c-Myc that regulate signalling pathways (e.g., AMPK, mTOR). High expression of p53 and the pro-apoptotic proteins cytochrome c and caspases without induction of apoptosis points to functions for these proteins in promoting cell differentiation. These results clearly show that VDAC1 depletion similarly leads to a rewiring of cancer cell metabolism in breast and lung cancer and glioblastoma, regardless of origin or mutational status. This metabolic reprogramming results in cell growth arrest and inhibited tumour growth while encouraging cell differentiation, thus generating cells with decreased proliferation capacity. These results further suggest VDAC1 to be an innovative and markedly potent therapeutic target.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.