Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (183)

Search Parameters:
Keywords = VDAC2

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 4690 KiB  
Article
Genome-Wide Characterization of VDAC Gene Family in Soybean (Glycine max L.) and In Silico Expression Profiling in Response to Drought and Salt Stress
by Muhammad Muneeb Ullah, Muqadas Aleem, Muhammad Mudassar Iqbal, Awais Riaz and Ainong Shi
Plants 2025, 14(14), 2101; https://doi.org/10.3390/plants14142101 - 8 Jul 2025
Viewed by 280
Abstract
Soybean (Glycine max L.) is grown worldwide to obtain edible oil, livestock feed, and biodiesel. However, drought and salt stress are becoming serious challenges to global soybean cultivation as they retard the growth of soybean plants and cause significant yield losses. Voltage-dependent [...] Read more.
Soybean (Glycine max L.) is grown worldwide to obtain edible oil, livestock feed, and biodiesel. However, drought and salt stress are becoming serious challenges to global soybean cultivation as they retard the growth of soybean plants and cause significant yield losses. Voltage-dependent anion-selective channel (VDAC) proteins are well-known for their role in drought and salt tolerance in crop plants. In this study, we identified 111 putative VDAC genes randomly distributed in genomes of 14 plant species, including cultivated soybean (Glycine max) and wild soybean (Glycine soja). The comparative phylogenetic studies classified these genes into six different clades and found the highest structural similarities among VDAC genes of G. max and G. soja. From the conserved domain database, porin-3 (PF01459) was found to be the conserved domain in all VDAC proteins. Furthermore, gene annotation studies revealed the role of GmaVDAC proteins in voltage-gated anion channel activity. These proteins were also found to interact with other proteins, especially mitochondrial receptors. A total of 103 miRNAs were predicted to target fifteen GmaVDAC genes. In G. max, these genes were found to be segmentally duplicated and randomly distributed on twelve chromosomes. Transcriptomic analysis revealed that the GmaVDAC18.2 gene showed overexpression in root nodules, whereas the GmaVDAC9.1, GmaVDAC18.1, and GmaVDAC18.2 genes showed overexpression under drought and salt stress conditions. Full article
(This article belongs to the Special Issue Functional Genomics and Molecular Breeding of Crops—2nd Edition)
Show Figures

Figure 1

22 pages, 5020 KiB  
Article
Upregulated Hexokinase-2 in Airway Epithelium Regulates Apoptosis and Drives Inflammation in Asthma via Peptidylprolyl Isomerase F
by Zhen Tian, Hongyan Zheng, Yan Fan, Boyu Li, Zhenli Huang, Meijia Wang, Jixian Zhang, Jianping Zhao, Shanshan Wang and Jungang Xie
Cells 2025, 14(13), 1004; https://doi.org/10.3390/cells14131004 - 1 Jul 2025
Viewed by 404
Abstract
Hexokinase catalyzes the first rate-limiting step glycolysis. However, the roles of hexokinase 2 (HK2) in asthma remain incompletely understood. This study aimed to investigate metabolic alterations in asthma, focusing on the expression, function and regulation of HK2. In this study, non-targeted metabolomics analysis [...] Read more.
Hexokinase catalyzes the first rate-limiting step glycolysis. However, the roles of hexokinase 2 (HK2) in asthma remain incompletely understood. This study aimed to investigate metabolic alterations in asthma, focusing on the expression, function and regulation of HK2. In this study, non-targeted metabolomics analysis of 20 asthma patients and 15 healthy controls identified metabolic alterations in asthma, particularly in the glycolytic pathways. Consistently, HK2 expression was elevated in both asthma individuals and mice with allergic airway inflammation. Airway epithelium–specific HK2 knockdown and pharmacological inhibition with 2-deoxy-D-glucose (2-DG) significantly attenuated airway inflammation and hyperresponsiveness in mice induced by ovalbumin/ lipopolysaccharide. Mechanistic analyses demonstrated that HK2 regulates epithelial apoptosis and inflammation via interaction with peptidylprolyl isomerase F (PPIF), independent of voltage-dependent anion channel 1 (VDAC1). Asthma is associated with metabolic reprogramming, characterized by alterations in lipid and glucose metabolism. These findings establish HK2 plays a crucial role in asthma pathogenesis by promoting airway epithelial apoptosis and inflammation in asthma, suggesting its potential as a therapeutic target. Full article
Show Figures

Figure 1

35 pages, 8277 KiB  
Review
Is the Voltage-Dependent Anion Channel a Major Player in Neurodegenerative Diseases?
by Sebastian Neumann and Rolf Heumann
Int. J. Mol. Sci. 2025, 26(13), 6138; https://doi.org/10.3390/ijms26136138 - 26 Jun 2025
Viewed by 651
Abstract
The family of voltage-dependent anion channels (VDACs) comprises three isoforms (VDAC-1, VDAC-2, VDAC-3). VDACs have been extensively described as localised in the outer mitochondrial membrane where they are involved in the exchange of ions, metabolites, and ATP/ADP between mitochondria and cytosol. The VDAC [...] Read more.
The family of voltage-dependent anion channels (VDACs) comprises three isoforms (VDAC-1, VDAC-2, VDAC-3). VDACs have been extensively described as localised in the outer mitochondrial membrane where they are involved in the exchange of ions, metabolites, and ATP/ADP between mitochondria and cytosol. The VDAC interacts with disease-specific proteins and thus regulates the mitochondrial function and controls the cellular energy resources, explaining its involvement in cell death and apoptosis. In addition, VDAC-1 and -2 can also be found at other cellular locations such as in the sarcoplasmic reticulum, in the endoplasmic reticulum, as well as in the plasma membrane. Through single-channel pore regulation, oligomerisation, or changed expression levels the VDAC is involved in different neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, Amyotrophic lateral sclerosis, Huntington’s disease, and others. Here, we critically summarise current discussions about the VDAC as a common key player for these diseases. We suggest that the VDAC acts as a transmembrane multifunctional regulatory protein which might serve as a pharmacological target for the development of novel drugs against neurodegenerative diseases such as the application of recombinant antibody technology. Full article
Show Figures

Figure 1

19 pages, 1176 KiB  
Article
Temporal Changes in Mitochondria-Centric Excitotoxic Responses Following Severe Penetrating Traumatic Brain Injury
by Hiren R. Modi, Sudeep Musyaju, Anke H. Scultetus and Jignesh D. Pandya
Biomedicines 2025, 13(7), 1520; https://doi.org/10.3390/biomedicines13071520 - 21 Jun 2025
Viewed by 347
Abstract
Background/Objectives: Traumatic brain injury (TBI) remains a significant and urgent medical concern for the US military. TBI triggers excitotoxic responses immediately, involving mitochondrial dysfunction characterized by loss of calcium (Ca2+) cycling, membrane damage and increased cell death. However, a comprehensive understanding [...] Read more.
Background/Objectives: Traumatic brain injury (TBI) remains a significant and urgent medical concern for the US military. TBI triggers excitotoxic responses immediately, involving mitochondrial dysfunction characterized by loss of calcium (Ca2+) cycling, membrane damage and increased cell death. However, a comprehensive understanding of mitochondria-centric excitotoxic responses over time has yet to be fully demonstrated after severe TBI. The current study evaluated mitochondria-centric time course responses between 30 min and 2 weeks (seven time points) after penetrating TBI (pTBI). Methods: Anesthetized adult male Sprague-Dawley rats were subjected to either 10% unilateral pTBI or Sham craniectomy. Animals were euthanized at various time points, and mitochondria were isolated from the injury core. Results: Post-injury mitochondrial Ca2+ homeostasis was significantly compromised in pTBI compared to the Sham group. In parallel, mitochondrial membrane integrity markers, including cytochrome c (Cyt C) and voltage-dependent anion channel (VDAC), showed significant reduction over time post-pTBI. Apoptosis-responsive markers, such as glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and B-cell lymphoma 2 (Bcl-2), exhibited elevated responses over time post-pTBI. Conclusions: Our results demonstrate profound insights into elevated excitotoxic mitochondrial damage after severe TBI. This time course study uncovers novel mitochondrial targets involved in TBI excitotoxicity and offers mitigation opportunities to alleviate excitotoxic responses after penetrating TBI. Full article
Show Figures

Figure 1

16 pages, 5130 KiB  
Article
Induced Mitophagy Promotes Cell Cycle Re-Entry in Adult Cardiomyocytes
by Rafeeq P. H. Ahmed, Onur Kanisicak and Perwez Alam
Cells 2025, 14(12), 853; https://doi.org/10.3390/cells14120853 - 6 Jun 2025
Viewed by 650
Abstract
Background: The limited regenerative capacity of adult mammalian cardiomyocytes (CMs) poses a significant challenge for cardiac repair following myocardial infarction. In contrast to adult mammals, CMs in zebrafish and newt hearts retain a lifelong capacity for proliferation and cardiac regeneration. Likewise, neonatal [...] Read more.
Background: The limited regenerative capacity of adult mammalian cardiomyocytes (CMs) poses a significant challenge for cardiac repair following myocardial infarction. In contrast to adult mammals, CMs in zebrafish and newt hearts retain a lifelong capacity for proliferation and cardiac regeneration. Likewise, neonatal mice exhibit a brief postnatal period, during which CMs retain the ability to proliferate and contribute to myocardial repair, which markedly diminishes within the first week of life. Emerging evidence indicates that adult CM cell cycle progression is critically influenced by oxidative stress. Adult mammalian CMs possess a high mitochondrial content to meet their substantial energy demands. However, this also leads to elevated reactive oxygen species (ROS) production, resulting in DNA damage and subsequent cell cycle arrest. We hypothesize that reducing the mitochondrial content in adult CMs will mitigate ROS production, thereby facilitating cell cycle progression. Methods: Adult CMs were isolated from adult rats (≥12 weeks old). To induce mitophagy, adult CMs were transfected with parkin-expressing plasmid and then treated with carbonyl cyanide 3-chlorophenylhydrazone (CCCP), a mitochondrial protonophore, for 7 days. Post-treatment assessments included the quantification of adult CM proliferation, mitochondrial content, and ROS levels. Results: CCCP-treated adult CMs exhibited a significant increase in proliferation markers, including EdU incorporation, KI67, phospho-histone H3, and Aurora B. Furthermore, CCCP treatment significantly reduced the mitochondrial content, as evidenced by decreased MitoTracker, TMRM, and Tom20 staining compared to controls. This was accompanied by electron microscopy analysis, which showed a significant reduction in the mitochondrial number in the adult CM after CCCP treatment. Moreover, our results also demonstrate a marked reduction in oxidative stress, demonstrated by lower 123-dihydro-rhodamine (123-DHR), CellROX signals, and VDAC. Conclusions: Our findings demonstrate that CCCP-mediated mitochondrial depletion reduces oxidative stress and promotes cell cycle re-entry in adult CM. This study provides direct experimental evidence and substantiates the role of elevated mitochondria and ROS levels in adult CM cell cycle exit. Full article
(This article belongs to the Section Cells of the Cardiovascular System)
Show Figures

Figure 1

19 pages, 5125 KiB  
Article
VDAC2 Mediates the Apoptosis of Cashmere Goat Hair Follicle Stem Cells Through the P53 Signaling Pathway
by Long Zhu, Yueqi Zhao, Mei Zhou, Xiaotong Guo, Yinxian Zhang, Dongjun Liu and Xudong Guo
Animals 2025, 15(11), 1671; https://doi.org/10.3390/ani15111671 - 5 Jun 2025
Viewed by 370
Abstract
Hair follicle stem cells (HFSCs) are pluripotent stem cells located in the bulges of hair follicles. Apoptosis regulates tissue homeostasis by eliminating unnecessary or damaged cells during development and aging. VDAC2, located in the outer mitochondrial membrane (MOM), is a key apoptosis regulator, [...] Read more.
Hair follicle stem cells (HFSCs) are pluripotent stem cells located in the bulges of hair follicles. Apoptosis regulates tissue homeostasis by eliminating unnecessary or damaged cells during development and aging. VDAC2, located in the outer mitochondrial membrane (MOM), is a key apoptosis regulator, but its role in cashmere goat hair follicles remains unclear. In previous studies, through proteomic sequencing, we found that VDAC2 was significantly differentially expressed in the anagen, catagen, and telogen phases of the hair follicles of Albas cashmere goats. This study aimed to explore the role of VDAC2 in secondary hair follicle stem cells (SHFSCs) and preliminarily investigate its regulatory mechanism through RNA-seq. Overexpression of VDAC2 promoted apoptosis in SHFSCs, while knockdown had the opposite effect. RNA-seq analysis, together with expression validation of downstream genes, indicates that the P53 signaling pathway may be involved in VDAC2-mediated SHFSC regulation. RT-qPCR and Western blotting confirmed that VDAC2 activated the P53 signaling pathway in SHFSCs. Furthermore, the use of a P53 inhibitor after VDAC2 overexpression partially rescued the apoptosis of cells caused by VDAC2. These results demonstrate that VDAC2 plays an important role in SHFSC apoptosis. Our findings greatly enhance our understanding of the role of VDAC2 in SHFSC apoptosis and hair follicle growth. Full article
(This article belongs to the Section Animal Physiology)
Show Figures

Figure 1

26 pages, 4306 KiB  
Article
Metformin-Induced Apoptosis Is Mediated Through Mitochondrial VDAC1
by Anna Shteinfer-Kuzmine, Meital M. Moyal, Aditya Karunanithi Nivedita, Sweta Trishna, Almog Nadir, Shubhandra Tripathi and Varda Shoshan-Barmatz
Pharmaceuticals 2025, 18(5), 757; https://doi.org/10.3390/ph18050757 - 20 May 2025
Viewed by 608
Abstract
Background: Besides diabetes mellitus, metformin has been identified as a potential therapeutic agent for treating various other conditions that include various cancers, cardiovascular diseases, neurodegenerative diseases, and aging. In cancer, metformin increased apoptotic cell death, while inhibiting it in neurodegenerative diseases. Thus, different [...] Read more.
Background: Besides diabetes mellitus, metformin has been identified as a potential therapeutic agent for treating various other conditions that include various cancers, cardiovascular diseases, neurodegenerative diseases, and aging. In cancer, metformin increased apoptotic cell death, while inhibiting it in neurodegenerative diseases. Thus, different modes of metformin action at the molecular level have been proposed. Methods: In this study, we present the mitochondria and the VDAC1 (voltage-dependent anion channel) as a potential target of metformin. Results: Metformin induces VDAC1 overexpression, its oligomerization, and subsequent apoptosis. Metformin analogs phenformin and buformin at much lower concentrations also induce VDAC1 overexpression, oligomerization, and cell death. We demonstrate the interaction of metformin with purified VDAC1, which inhibited its channel conduction in a voltage-dependent manner. Metformin bound to the synthetic VDAC1-N-terminal peptide and binding to this domain was also found by its molecular docking with VDAC1. Moreover, we demonstrated metformin binding to purified hexokinases (HK-I) with a 400-fold lower metformin concentration than that required for cell death induction. In cells, metformin induced HK-I detachment from the mitochondrial VDAC1. Lastly, metformin increased the expression of NLRP3 and ASC and induced their co-localization, suggesting inflammasome activation. Conclusions: The results suggest that VDAC1 is a target for metformin and its analogs, and this is associated with metformin’s adverse effects on many diseases. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

21 pages, 14030 KiB  
Article
Impact of Type 1 Diabetes on Testicular Microtubule Dynamics, Sperm Physiology, and Male Reproductive Health in Rat
by Alessandra Biasi, Maria Rosaria Ambruosi, Maria Zelinda Romano, Serena Boccella, Sara Falvo, Francesca Guida, Francesco Aniello, Sabatino Maione, Massimo Venditti and Sergio Minucci
Int. J. Mol. Sci. 2025, 26(10), 4579; https://doi.org/10.3390/ijms26104579 - 10 May 2025
Viewed by 681
Abstract
Type 1 diabetes (T1D) is a chronic metabolic disease defined by sustained hyperglycemia, leading to oxidative stress (OS) and systemic complications, including male subfertility. This study investigates the potential impact of T1D-induced OS on microtubule (MTs) dynamics and microtubule-associated proteins (MAPs) in the [...] Read more.
Type 1 diabetes (T1D) is a chronic metabolic disease defined by sustained hyperglycemia, leading to oxidative stress (OS) and systemic complications, including male subfertility. This study investigates the potential impact of T1D-induced OS on microtubule (MTs) dynamics and microtubule-associated proteins (MAPs) in the testis and spermatozoa (SPZ). Using a streptozotocin-induced T1D rat model, we examined the expression and localization of key MAPs, including Microtubule Affinity-Regulating Kinase 4 (MARK4), Microtubule-Associated Protein 1A (MAP1A), Dynein Light Chain LC8-Type 1 (DYNLL1), Prolyl Endopeptidase (PREP), and Radial Spoke Head 6 Homolog A (RSPH6A), alongside sperm functional parameters. Our findings showed that T1D significantly impaired the expression and distribution of these proteins, which may affect MTs organization and be associated with cytoskeletal disorganization, and impaired germ cell differentiation. Moreover, T1D rats exhibited reduced sperm count, viability, and motility, accompanied by increased DNA fragmentation and chromatin defects. Elevated levels of 4-hydroxy-2-nonenal (4-HNE), a marker of OS, were detected in SPZ, particularly in the acrosome and flagellum, correlating with mitochondrial dysfunction and ATP depletion. Additionally, decreased intracellular Ca2+ levels, downregulation of Cation Channel of Sperm (CATSPER) and Voltage-Dependent Anion Channel 3 (VDAC3), and altered tubulin acetylation, possibly due to imbalanced Alpha-Tubulin N-Acetyltransferase 1 (ATAT1) and Histone Deacetylase 6 (HDAC6) expression, were also associated with impaired sperm motility. The combined data suggest that T1D-induced OS is linked to disrupted MTs dynamics, which may contribute to testicular dysfunction and reduced sperm quality, potentially affecting male fertility. A better understanding of these associations may support the development of therapeutic strategies to mitigate the reproductive consequences of T1D and improve male fertility outcomes. Full article
Show Figures

Graphical abstract

24 pages, 7713 KiB  
Article
Resveratrol’s Pro-Apoptotic Effects in Cancer Are Mediated Through the Interaction and Oligomerization of the Mitochondrial VDAC1
by Tal Raviv, Anna Shteinfer-Kuzmine, Meital M. Moyal and Varda Shoshan-Barmatz
Int. J. Mol. Sci. 2025, 26(9), 3963; https://doi.org/10.3390/ijms26093963 - 22 Apr 2025
Viewed by 867
Abstract
Resveratrol is a naturally occurring phenolic compound found in various foods such as red wine, chocolate, peanuts, and blueberries. Both in-vitro and in-vivo studies have shown that it has a broad spectrum of pharmacological effects such as providing cellular protection and promoting longevity. [...] Read more.
Resveratrol is a naturally occurring phenolic compound found in various foods such as red wine, chocolate, peanuts, and blueberries. Both in-vitro and in-vivo studies have shown that it has a broad spectrum of pharmacological effects such as providing cellular protection and promoting longevity. These effects include antioxidant, anti-inflammatory, neuroprotective, and anti-viral properties, as well as improvements in cardio-metabolic health and anti-aging benefits. Additionally, resveratrol has demonstrated the ability to induce cell death and inhibit tumor growth across different types and stages of cancer. However, the dual effects of resveratrol—acting to support cell survival in some contexts, while inducing cell death in others—is still not fully understood. In this study, we identify a novel target for resveratrol: the voltage-dependent anion channel 1 (VDAC1), a multi-functional outer mitochondrial membrane protein that plays a key role in regulating both cell survival and death. Our findings show that resveratrol increased VDAC1 expression levels and promoted its oligomerization, leading to apoptotic cell death. Additionally, resveratrol elevated intracellular Ca2+ levels and enhanced the production of reactive oxygen species (ROS). Resveratrol also induced the detachment of hexokinase I from VDAC1, a key enzyme in metabolism, and regulating apoptosis. When VDAC1 expression was silenced using specific siRNA, resveratrol-induced cell death was significantly reduced, indicating that VDAC1 is essential for its pro-apoptotic effects. Additionally, both resveratrol and its analog, trans-2,3,5,4′-tetrahydroxystilbene-2-O-glucoside (TSG), directly interacted with purified VDAC1, as revealed by microscale thermophoresis, with similar binding affinities. However, unlike resveratrol, TSG did not induce VDAC1 overexpression or apoptosis. These results demonstrate that resveratrol-induced apoptosis is linked to increased VDAC1 expression and its oligomerization. This positions resveratrol not only as a protective agent, but also as a pro-apoptotic compound. Consequently, resveratrol offers a promising therapeutic approach for cancer, with potentially fewer side effects compared to conventional treatments, due to its natural origins in plants and food products. Full article
(This article belongs to the Collection Feature Papers in Molecular Oncology)
Show Figures

Figure 1

26 pages, 8882 KiB  
Article
Effect of 1-DNJ on Oxidative Stress-Induced Apoptosis in Porcine Ovarian GCs Through Modulation of the PERK-ATF4/MFN2 Signaling Pathway
by Wenwen Xing, Mengxuan Li, Binbin Wang, Lele Huo, Wanru Tian, Fangcai Ge, Manman Shen, Liumei Sun, Jiying Liu and Shali Yu
Antioxidants 2025, 14(4), 456; https://doi.org/10.3390/antiox14040456 - 11 Apr 2025
Viewed by 774
Abstract
Oxidative stress (OS) is regarded as a major contributor to granulosa cellapoptosis in ovarian disease. 1-Deoxynojirimycin (1-DNJ), a naturally occurring plant alkaloid, exhibits antioxidant, anti-inflammatory, and metabolism-modulating properties. Mitochondria and endoplasmic reticulum (ER), crucial organelles regulating oxidative balance, interact through mitochondria-associated endoplasmic reticulum [...] Read more.
Oxidative stress (OS) is regarded as a major contributor to granulosa cellapoptosis in ovarian disease. 1-Deoxynojirimycin (1-DNJ), a naturally occurring plant alkaloid, exhibits antioxidant, anti-inflammatory, and metabolism-modulating properties. Mitochondria and endoplasmic reticulum (ER), crucial organelles regulating oxidative balance, interact through mitochondria-associated endoplasmic reticulum membranes (MAMs) for signaling and molecular exchange. However, it remains unclear whether 1-DNJ attenuates oxidative damage in ovarian granulosa cells (GCs) via MAMs-mediated ER–mitochondria crosstalk, which needs further exploration. This study aimed to investigate the mechanisms by which 1-DNJ affects oxidative damage and apoptosis induced by OS in porcine follicular GCs by regulating mitochondrial function, MAMs, and ER interactions. Here, we found that GCs suffered from OS, accompanied by the up-regulation of ROS and MDA, alongside reduced activity of antioxidant enzymes (CAT and T-SOD). Further studies revealed that the up-regulation of MAMs proteins (MFN2, MCU, and VDAC1) and pro-apoptosis proteins (BAX and Cleaved-capase3), along with increased mitochondrial ROS and Ca2+ levels, led to the down-regulation of MMP and ATP content. These, in turn, triggered mitochondrial dysfunction, and MAMs destabilization, and subsequent apoptosis. Additionally, the up-regulation of the protein levels of P-PERK/PERK, GRP78, ATF4, and CHOP protein expression activated the PERK-ATF4 signaling pathway, which triggered endoplasmic reticulum stress (ERS). Conversely, 1-DNJ alleviated H2O2-induced mitochondrial and MAMs dysfunction and ERS, which in turn attenuated apoptosis. Further, ATF4 knockdown inhibited MFN2 protein expression, which attenuated H2O2-induced MMP inhibition, Ca2+ overload, ROS production, and mitochondrial damage. In summary, 1-DNJ mitigated OS-induced mitochondrial dysfunction in GCs and regulated ER–mitochondrial communication through MAMs, reducing OS-induced apoptosis. The present study demonstrates that 1-DNJ protects ovarian GCs from OS-induced damage by modulating ER and mitochondrial homeostasis through MAMs, offering new perspectives and a theoretical basis for the treatment of ovarian diseases. Full article
Show Figures

Figure 1

16 pages, 3697 KiB  
Article
D-M159 Synergistically Induces Apoptosis in HeLa Cells Through Endoplasmic Reticulum Stress and Mitochondrial Dysfunction
by Yuanyuan Li, Dingding Li, Zonghan Jiang, Zhihang Yuan, Zhiliang Sun and Leisheng Sun
Int. J. Mol. Sci. 2025, 26(7), 3172; https://doi.org/10.3390/ijms26073172 - 29 Mar 2025
Viewed by 801
Abstract
Pore-forming peptides are promising antimicrobial and anticancer agents due to their membrane selectivity and biodegradability. Our prior work identified peptide M159, which permeabilized synthetic phosphatidylcholine liposomes without mammalian cell toxicity. Here, we report that the D-type variant (D-M159) induces apoptosis in HeLa cells [...] Read more.
Pore-forming peptides are promising antimicrobial and anticancer agents due to their membrane selectivity and biodegradability. Our prior work identified peptide M159, which permeabilized synthetic phosphatidylcholine liposomes without mammalian cell toxicity. Here, we report that the D-type variant (D-M159) induces apoptosis in HeLa cells under starvation. To explore its anticancer mechanism, we analyzed D-M159 cytotoxicity, intracellular uptake, and apoptotic pathways via flow cytometry, confocal microscopy, and Western blot. Calcium dynamics and mitochondrial function were examined via specific labeling and functional assays. Results revealed that D-M159 exhibited starvation-dependent, dose-responsive cytotoxicity and triggered apoptosis in HeLa cells. Mechanistic studies indicated that D-M159 entered the cells via caveolin-dependent and caveolae-dependent endocytosis pathways and induced endoplasmic reticulum stress in HeLa cells by up-regulating proteins such as ATF6, p-IRE1, PERK, GRP78, and CHOP. Meanwhile, D-M159 promoted the expression of IP3R1, GRP75, and VDAC1, which led to mitochondrial calcium iron overload, decreased mitochondrial membrane potential, and increased reactive oxygen species (ROS) generation, thereby activating the mitochondrial apoptotic pathway and inducing the aberrant expression of Bax, Bcl-2, Caspase-9, and Caspase-3. This study showed that D-M159 synergistically induced apoptosis in starved HeLa cells through endoplasmic reticulum stress and mitochondrial dysfunction, demonstrating its potential as a novel anticancer agent. Full article
(This article belongs to the Section Macromolecules)
Show Figures

Figure 1

26 pages, 5820 KiB  
Article
MIRO1 Is Required for Dynamic Increases in Mitochondria-ER Contact Sites and Mitochondrial ATP During the Cell Cycle
by Benney T. Endoni, Olha M. Koval, Chantal Allamargot, Tara Kortlever, Lan Qian, Riley J. Sadoski, Denise Juhr and Isabella M. Grumbach
Cells 2025, 14(7), 482; https://doi.org/10.3390/cells14070482 - 22 Mar 2025
Cited by 1 | Viewed by 1072
Abstract
Mitochondria-ER contact sites (MERCS) are vital for mitochondrial dynamics, lipid exchange, Ca2+ homeostasis, and energy metabolism. We examined whether mitochondrial metabolism changes during the cell cycle depend on MERCS dynamics and are regulated by the outer mitochondrial protein mitochondrial rho GTPase 1 [...] Read more.
Mitochondria-ER contact sites (MERCS) are vital for mitochondrial dynamics, lipid exchange, Ca2+ homeostasis, and energy metabolism. We examined whether mitochondrial metabolism changes during the cell cycle depend on MERCS dynamics and are regulated by the outer mitochondrial protein mitochondrial rho GTPase 1 (MIRO1). Wound healing was assessed in mice with fibroblast-specific deletion of MIRO1. Wild-type and MIRO1-/- fibroblasts and vascular smooth muscle cells were evaluated for proliferation, cell cycle progression, number of MERCS, distance, and protein composition throughout the cell cycle. Restoration of MIRO1 mutants was used to test the role of MIRO1 domains; Ca2+ transients and mitochondrial metabolism were evaluated using biochemical, immunodetection, and fluorescence techniques. MERCS increased in number during G1/S compared with during G0, which was accompanied by a notable rise in protein–protein interactions involving VDAC1 and IP3R as well as GRP75 and MIRO1 by proximity-ligation assays. Split-GFP ER/mitochondrial contacts of 40 nm also increased. Mitochondrial Ca2+ concentration ([Ca2+]), membrane potential, and ATP levels correlated with the formation of MERCS during the cell cycle. MIRO1 deficiency blocked G1/S progression and the cell-cycle-dependent formation of MERCS and altered ER Ca2+ release and mitochondrial Ca2+ uptake. MIRO1 mutants lacking the Ca2+-sensitive EF hands or the transmembrane domain did not rescue cell proliferation or the formation of MERCS. MIRO1 controls an increase in the number of MERCS during cell cycle progression and increases mitochondrial [Ca2+], driving metabolic activity and proliferation through its EF hands. Full article
Show Figures

Figure 1

19 pages, 2340 KiB  
Article
Ferroptosis-Related Genes as Molecular Markers in Bovine Mammary Epithelial Cells Challenged with Staphylococcus aureus
by Yue Xing, Siyuan Mi, Gerile Dari, Zihan Zhang, Siqian Chen and Ying Yu
Int. J. Mol. Sci. 2025, 26(6), 2506; https://doi.org/10.3390/ijms26062506 - 11 Mar 2025
Cited by 1 | Viewed by 845
Abstract
Staphylococcus aureus-induced mastitis is a significant cause of economic losses in the dairy industry, yet its molecular mechanisms remain poorly defined. Although ferroptosis, a regulated cell death process, is associated with inflammatory diseases, its role in bovine mastitis is unknown. In this [...] Read more.
Staphylococcus aureus-induced mastitis is a significant cause of economic losses in the dairy industry, yet its molecular mechanisms remain poorly defined. Although ferroptosis, a regulated cell death process, is associated with inflammatory diseases, its role in bovine mastitis is unknown. In this study, 11 S. aureus strains were isolated from milk samples obtained from cows with clinical or subclinical mastitis. Transcriptome analysis of Mac-T cells challenged with isolated S. aureus identified differentially expressed genes (DEGs). Enrichment analysis revealed significant associations between DEG clusters and traits related to bovine mastitis. KEGG pathway enrichment revealed ferroptosis, Toll-like receptor, and TNF signaling as significantly enriched pathways. Weighted gene co-expression network analysis (WGCNA) further prioritized ferroptosis-related genes (HMOX1, SLC11A2, STEAP3, SAT1, and VDAC2) involved in iron metabolism. Notably, the expression levels of HMOX1 and SAT1 were significantly increased in S. aureus-challenged Mac-T cells, and this upregulation was consistent with trends observed in transcriptome data from mother–daughter pairs of cows with subclinical mastitis caused by S. aureus infection. Furthermore, Ferrostatin-1 treatment significantly reduced the expression of HMOX1 and SAT1 in S. aureus-challenged cells, confirming the involvement of ferroptosis in this process. This study reveals that ferroptosis plays a key role in S. aureus-induced mastitis and highlights its potential as a target for molecular breeding strategies aimed at improving bovine mastitis resistance. Full article
Show Figures

Figure 1

21 pages, 3475 KiB  
Review
VDAC1: A Key Player in the Mitochondrial Landscape of Neurodegeneration
by Shirel Argueti-Ostrovsky, Shir Barel, Joy Kahn and Adrian Israelson
Biomolecules 2025, 15(1), 33; https://doi.org/10.3390/biom15010033 - 30 Dec 2024
Cited by 2 | Viewed by 2470
Abstract
Voltage-Dependent Anion Channel 1 (VDAC1) is a mitochondrial outer membrane protein that plays a crucial role in regulating cellular energy metabolism and apoptosis by mediating the exchange of ions and metabolites between mitochondria and the cytosol. Mitochondrial dysfunction and oxidative stress are central [...] Read more.
Voltage-Dependent Anion Channel 1 (VDAC1) is a mitochondrial outer membrane protein that plays a crucial role in regulating cellular energy metabolism and apoptosis by mediating the exchange of ions and metabolites between mitochondria and the cytosol. Mitochondrial dysfunction and oxidative stress are central features of neurodegenerative diseases. The pivotal functions of VDAC1 in controlling mitochondrial membrane permeability, regulating calcium balance, and facilitating programmed cell death pathways, position it as a key determinant in the delicate balance between neuronal viability and degeneration. Accordingly, increasing evidence suggests that VDAC1 is implicated in the pathophysiology of neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and others. This review summarizes the current findings on the contribution of VDAC1 to neurodegeneration, focusing on its interactions with disease-specific proteins, such as amyloid-β, α-synuclein, and mutant SOD1. By unraveling the complex involvement of VDAC1 in neurodegenerative processes, this review highlights potential avenues for future research and drug development aimed at alleviating mitochondrial-related neurodegeneration. Full article
Show Figures

Figure 1

16 pages, 4153 KiB  
Article
Metabolic Activity in Human Intermuscular Adipose Tissue Directs the Response of Resident PPARγ+ Macrophages to Fatty Acids
by Xiaoying Chen, Sebastian Ludger Schubert, Aline Müller, Miguel Pishnamaz, Frank Hildebrand and Mahtab Nourbakhsh
Biomedicines 2025, 13(1), 10; https://doi.org/10.3390/biomedicines13010010 - 25 Dec 2024
Viewed by 4338
Abstract
Background/Objectives: Peroxisome proliferator-activated receptor gamma (PPARγ) is a fatty acid-binding transcription activator of the adipokine chemerin. The key role of PPARγ in adipogenesis was established by reports on adipose tissue-resident macrophages that express PPARγ. The present study examined PPARγ+ macrophages in [...] Read more.
Background/Objectives: Peroxisome proliferator-activated receptor gamma (PPARγ) is a fatty acid-binding transcription activator of the adipokine chemerin. The key role of PPARγ in adipogenesis was established by reports on adipose tissue-resident macrophages that express PPARγ. The present study examined PPARγ+ macrophages in human skeletal muscle tissues, their response to fatty acid (FA) species, and their correlations with age, obesity, adipokine expression, and an abundance of other macrophage phenotypes. Methods: An ex vivo human skeletal muscle model with surgical specimens that were maintained without or with FAs for up to 11 days was utilized. Immunofluorescence analysis was used to detect macrophage phenotypes and mitochondrial activity. Preconfigured arrays were used to detect the expression of 34 different adipokines and chemokines. Results: Data from 14 adults revealed that PPARγ+ macrophages exclusively reside in intermuscular adipose tissue (IMAT), and their abundance correlates with the metabolic status of surrounding adipocytes during tissue maintenance in vitro for 9–11 days. Elevated fatty acid levels lead to significant increases in PPARγ+ populations, which are correlated with the donor’s body mass index (BMI). Conclusions: PPARγ+ macrophages represent a distinctly specialized population of regulatory cells that reside within human IMATs in accordance with their metabolic status. Thus, future in-depth studies on IMAT-resident PPARγ+ macrophage action mechanisms will elucidate the role of skeletal muscle in the pathogenesis of human metabolic dysfunction. Full article
(This article belongs to the Special Issue The Role of Chemerin in Human Disease2nd Edition)
Show Figures

Figure 1

Back to TopTop