1
|
Gong Y, Zhao M, Pan M, Zhao Y, Liu J, Wen H, Wang J. Harmine derivative H-2-168 induces the death of Echinococcus granulosus by regulating mitochondrial fusion and fission. PHARMACEUTICAL BIOLOGY 2025; 63:188-200. [PMID: 40188381 PMCID: PMC11980216 DOI: 10.1080/13880209.2025.2485898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/08/2025]
Abstract
CONTEXT H-2-168 has pharmacological effects similar to those of harmine, with less toxicity. The health of cells and organisms depends on a delicate balance between mitochondrial fusion and fission. OBJECTIVE This study investigated the roles of H-2-168 and mitochondrial fusion and fission in Echinococcus granulosus. MATERIALS AND METHODS Notably, E. granulosus were isolated from fresh sheep livers, and then treated with H-2-168 (25 μg/mL), mitochondrial division inhibitor 1 (Mdivi-1, 25 μg/mL) or the combination of H-2-168:Mdivi-1 (25 μg/mL:12.5 μg/mL). After 24 h of culture, the indices related to E. granulosus were measured. Additionally, Drp1 was knocked down to explore its effects on E. granulosus growth. RESULTS The EC50 values of H-2-168, Mdivi-1 and H-2-168:Mdivi-1 against E. granulosus were 44.171, 117.882 and 32.924 μg/mL, respectively. Compared with H-2-168 or Mdivi-1, the combination of H-2-168 and Mdivi-1 showed better inhibitory effects on E. granulosus viability, as well as increased levels of ROS and LDH, decreased ATP levels, inhibited mitochondrial activity and reduced mitochondrial membrane potential (p < 0.05), with the upregulation of Caspase-3, Cyt-c, Drp1, Fis1 and downregulation of Bcl-2, Mfn2 and OPA1. Additionally, Drp1 knockdown was successfully performed in E. granulosus, which significantly inhibited E. granulosus viability (p < 0.05) and further downregulated Mfn2 expression induced by H-2-168. DISCUSSION AND CONCLUSION Drp1 is closely associated with mitochondrial fusion and fission, and H-2-168 may promote E. granulosus death through disrupting the balance between mitochondrial fusion and fission.
Collapse
Affiliation(s)
- Yuehong Gong
- Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China
| | - Meiling Zhao
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Meichi Pan
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Yicong Zhao
- Department of Pharmacognosy, School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Junpeng Liu
- Department of Medicine, School of Pharmacy, Xinjiang Medical University, Urumqi, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jianhua Wang
- Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Clinical Drug Research, Urumqi, China
| |
Collapse
|
2
|
Lee JS, Kao DJ, Worledge CS, Villamaria ZF, Wang RX, Welch NM, Kostelecky RE, Colgan SP. E. coli genetically modified for purine nucleobase release promotes butyrate generation and colonic wound healing during DSS insult. Gut Microbes 2025; 17:2490211. [PMID: 40247632 PMCID: PMC12013446 DOI: 10.1080/19490976.2025.2490211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 12/27/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
The gut microbiota transforms energy stored as undigestible carbohydrates into a remarkable number of metabolites that fuel intestinal bacterial communities and the host tissue. Colonic epithelial cells at the microbiota-host interface depend upon such microbiota-derived metabolites (MDMs) to satisfy their energy requisite. Microbial dysbiosis eliciting MDM loss contributes to barrier dysfunction and mucosal disease. Recent work has identified a role for microbiota-sourced purines (MSPs), notably hypoxanthine, as an MDM salvaged by the colonic epithelium for nucleotide biogenesis and energy balance. Here, we investigated the role of MSPs in mice during disease-modeled colonic energetic stress using a strain of E. coli genetically modified for enhanced purine nucleobase release (E. coli Mutant). E. coli Mutant colonization protected against DSS-induced tissue damage and permeability while promoting proliferation for wound healing. Metabolite and metagenomic analyses suggested a colonic butyrate-purine nucleobase metabolic axis, wherein the E. coli Mutant provided purine substrate for Clostridia butyrate production and host purine salvage, altogether supplying the host substrate for efficient nucleotide biogenesis and energy balance.
Collapse
Affiliation(s)
- J. Scott Lee
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Daniel J. Kao
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Corey S. Worledge
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Zachary F. Villamaria
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Ruth X. Wang
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Nichole M. Welch
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| | - Rachael E. Kostelecky
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| |
Collapse
|
3
|
Pradeepkiran JA, Islam MA, Sehar U, Reddy AP, Vijayan M, Reddy PH. Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 2025; 108:102734. [PMID: 40120948 DOI: 10.1016/j.arr.2025.102734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/26/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide. It is characterized by the accumulation of beta-amyloid and phosphorylated tau, synaptic damage, and mitochondrial abnormalities in the brain, leading to the progressive loss of cognitive function and memory. In AD, emerging research suggests that lifestyle factors such as a healthy diet and regular exercise may play a significant role in delaying the onset and progression of the disease. Mitochondria are often referred to as the powerhouse of the cell, as they are responsible for producing the energy to cells, including neurons to maintain cognitive function. Our article elaborates on how mitochondrial quality and function decline with age and AD, leading to an increase in oxidative stress and a decrease in ATP production. Decline in mitochondrial quality can impair cellular functions contributing to the development and progression of disease with the loss of neuronal functions in AD. This article also covered mitophagy, the process by which damaged or dysfunctional mitochondria are selectively removed from the cell to maintain cellular homeostasis. Impaired mitophagy has been implicated in the progression and pathogenesis of AD. We also discussed the impact of impaired mitophagy implicated in AD, as the accumulation of damaged mitochondria can lead to increased oxidative stress. We expounded how dietary interventions and exercise can help to improve mitochondrial quality, and mitochondrial function and enhance mitophagy in AD. A diet rich in antioxidants, polyphenols, and mitochondria-targeted small molecules has been shown to enhance mitochondrial function and protect against oxidative stress, particularly in neurons with aged and mild cognitively impaired subjects and AD patients. Promoting a healthy lifestyle, mainly balanced diet and regular exercise that support mitochondrial health, in an individual can potentially delay the onset and progression of AD. In conclusion, a healthy diet and regular exercise play a crucial role in maintaining mitochondrial quality and mitochondrial function, in turn, enhancing mitophagy and synaptic activities that delay AD in the elderly populations.
Collapse
Affiliation(s)
| | - Md Ariful Islam
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
4
|
Shi Y, Cao Z, Ge L, Lei L, Tao D, Zhong J, Xu D, Geng T, Li X, Li Z, Xing S, Wu X, Wang Z, Li L. Rotenone adaptation promotes migration and invasion of p53-wild-type colon cancer through lipid metabolism. Clin Transl Oncol 2025; 27:2557-2567. [PMID: 39612123 DOI: 10.1007/s12094-024-03785-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/10/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The association between mitochondrial dysfunction and multiple metabolic adaptations is increasingly being proven. We previously elucidated that mitochondrial complex I deficiency can promote glycolysis in mut-p53 SW480 cells. However, studies have revealed a phenotype with attenuated glycolysis but enhanced fatty acid oxidation (FAO) in invasive tumors. The interplay between complex I and FAO in carcinogenesis remains obscure. METHODS The p53 wild-type RKO cells were exposed to rotenone over at least 2 months to acquire rotenone adaptation cells. Then the transwell invasion assays and expression of metabolic enzymes were first detected in rotenone adaptation cells to illustrate whether rotenone adaptation is correlated with the invasion and FAO. The levels of epithelial-to-mesenchymal transition (EMT)-related proteins and acetyl-CoA in rotenone adaptation cells treated with etomoxir (ETO) and acetate were evaluated to verify the role of CPT1A in regulating invasion. Finally, the levels of reactive oxygen species (ROS) were detected. Meanwhile, the invasiveness and histone acetylation levels of rotenone adaptation cells were observed after adding an ROS inhibitor (N-acetyl-L-cysteine NAC) to demonstrate the molecular connection between FAO and invasion during rotenone adaptation. RESULTS We found long-term exposure to rotenone (a mitochondrial complex I inhibitor) led to EMT and high CPT1A expression in wt-p53 colon cancer. The inhibition of CPT1A suppressed the invasion and reduced histone acetylation, which was rescued by supplementing with acetate. Mechanistically, ROS is crucial for lipid metabolism remodeling. CONCLUSION Our study provides a novel understanding of the role of complex I in lipid reprogramming facilitating colon cancer invasion and metastasis.
Collapse
Affiliation(s)
- Yingying Shi
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Zhen Cao
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Ling Ge
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Lin Lei
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Dan Tao
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Juan Zhong
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Dan Xu
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Tao Geng
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Xuetao Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Ziwei Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Shuaishuai Xing
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Xinyu Wu
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Zhongxu Wang
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China
| | - Linjun Li
- Department of Oncology, Maternal and Child Health Hospital of Hubei Province, Wuhan, 430070, China.
| |
Collapse
|
5
|
D'Alessandro MCB, Kanaan S, Geller M, Praticò D, Daher JPL. Mitochondrial dysfunction in Alzheimer's disease. Ageing Res Rev 2025; 107:102713. [PMID: 40023293 DOI: 10.1016/j.arr.2025.102713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive decline and distinct neuropathological features. The absence of a definitive cure presents a significant challenge in neurology and neuroscience. Early clinical manifestations, such as memory retrieval deficits and apathy, underscore the need for a deeper understanding of the disease's underlying mechanisms. While amyloid-β plaques and tau neurofibrillary tangles have dominated research efforts, accumulating evidence highlights mitochondrial dysfunction as a central factor in AD pathogenesis. Mitochondria, essential cellular organelles responsible for energy production necessary for neuronal function become impaired in AD, triggering several cellular consequences. Factors such as oxidative stress, disturbances in energy metabolism, failures in the mitochondrial quality control system, and dysregulation of calcium release are associated with mitochondrial dysfunction. These abnormalities are closely linked to the neurodegenerative processes driving AD development and progression. This review explores the intricate relationship between mitochondrial dysfunction and AD pathogenesis, emphasizing its role in disease onset and progression, while also considering its potential as a biomarker and a therapeutic target.
Collapse
Affiliation(s)
- Maria Clara Bila D'Alessandro
- Universidade Federal Fluminense, Faculty of Medicine, Desembargador Athayde Parreiras road 100, Niterói, Rio de Janeiro, Brazil.
| | - Salim Kanaan
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| | - Mauro Geller
- Unifeso, Department of Immunology and Microbiology, Alberto Torres avenue 111, Teresópolis, Rio de Janeiro, Brazil
| | - Domenico Praticò
- Department of Neurosciences, Lewis Katz School of Medicine. Temple University, 3500 North Broad Street, Philadelphia, PA, United States.
| | - João Paulo Lima Daher
- Universidade Federal Fluminense, Faculty of Medicine, Department of Pathology, Marquês do Paraná road, 303, 2nd floor, Niterói, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Tian Z, Wang X, Luan Y, Liu R. TERT's protective effect on trophoblast cells: Reducing mitochondrial ROS. Reprod Toxicol 2025; 134:108899. [PMID: 40139512 DOI: 10.1016/j.reprotox.2025.108899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE The principal objective of this project is to elucidate the ameliorating function of telomerase reverse transcriptase (TERT) on placental trophoblast dysfunction and its associated mitochondrial dysfunction. METHODS We utilised 50 μM hydrogen peroxide (H2O2) to induce an oxidative stress model in HTR-8/SVneo cells (NC group and OS group), subsequently establishing three experimental conditions via lentiviral transfection: untreated controls (Con group), TERT-overexpressing cells (OE group), and empty vector-transfected negative cells (Neg group). In order to explore the role of mitochondrial TERT, we employed the Src kinase familyinhibitor PP1 (4-Amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine), a cell-permeable small molecule specifically blocking the translocation of TERT in HTR-8/SVneo cells with overexpressed TERT. We established four experimental conditions: untreated overexpression group (OE group), H2O2-stimulated group (OS group), co-treatment group of H2O2 and PP1 (PP1 OS group), and PP1 treatment group alone (PP1 group). The expression and localization of TERT were analysed via qRT-PCR, immunofluorescence, and Western blotting. In addition, the cellular functions (viability, migration, invasion) were assessed using CCK-8, Calcein AM/PI staining, wound healing assay and Transwell assay. Mitochondrial integrity was thoroughly assessed using a mitochondrial membrane potential assay, an ATP content assay, a mtDNA copy number quantification and a mitochondrial morphology assay. RESULTS The OE group demonstrated significantly reduced reactive oxygen species (ROS) levels compared to Con and Neg groups, accompanied by enhanced cellular viability and improved migratory and invasive capacities. Mitochondrial functional analyses revealed superior outcomes in the OE group, including stabilized mitochondrial membrane potential and ATP production, maintained mtDNA copy numbers, and preserved mitochondrial ultrastructure. Conversely, pharmacological inhibition with PP1 - a Src kinase family inhibitor that blocks TERT mitochondrial translocation - effectively abolished these protective effects. PP1-treated cells exhibited exacerbated oxidative stress, impaired cell viability, and diminished motility, confirming the essential role of mitochondrial-localized TERT in cellular homeostasis. CONCLUSIONS Elevated mitochondrial TERT levels were able to effectively mitigate the cellular functional damage induced by elevated ROS levels by maintaining mitochondrial stability. The above findings provide a theoretical basis for the development of intervention strategies for pregnancy-related disorders based on the regulation of mitochondrial TERT function.
Collapse
Affiliation(s)
- Zongyuan Tian
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Xueran Wang
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Yingyi Luan
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China
| | - Ruixia Liu
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100026, China.
| |
Collapse
|
7
|
Kong L, Li S, Fu Y, Cai Q, Zhai Z, Liang J, Ma T. Microplastics/nanoplastics contribute to aging and age-related diseases: Mitochondrial dysfunction as a crucial role. Food Chem Toxicol 2025; 199:115355. [PMID: 40020987 DOI: 10.1016/j.fct.2025.115355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/08/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
The pervasive utilization of plastic products has led to a significant escalation in plastic waste accumulation. Concurrently, the implications of emerging pollutants such as microplastics (MPs) and nanoplastics (NPs) on human health are increasingly being acknowledged. Recent research has demonstrated that MPs/NPs may contribute to the onset of human aging and age-related diseases. Additionally, MPs/NPs have the potential to induce mitochondrial damage, resulting in mitochondrial dysfunction. Mitochondrial dysfunction is widely recognized as a hallmark of aging; thus, it is necessary to elucidate the relationship between them. In this article, we first elucidate the distribution of MPs/NPs in various environmental media, their pathways into the human body, and their subsequent distribution within human tissues and organs. Subsequently, we examine the interplay between MPs/NPs, mitochondrial dysfunction, and the aging process. We aspire that this article will enhance awareness regarding the toxicity of MPs/NPs while also offering a theoretical framework to support the development of improved regulatory policies in the future.
Collapse
Affiliation(s)
- Liang Kong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Disease, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Shuhao Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Yu Fu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Qinyun Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Zhengyu Zhai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Disease, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Disease, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| |
Collapse
|
8
|
Zhao M, Xie X, Ding Y, Zhang Q, Liu X, Lin T, Lan L, Hong G. Glabridin protects against paraquat-induced acute lung injury by targeting ME1 to mitigate oxidative stress, mitochondrial dysfunction, and cGAS-STING activation. Free Radic Biol Med 2025; 235:317-334. [PMID: 40316060 DOI: 10.1016/j.freeradbiomed.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/30/2025] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Acute lung injury (ALI) resulting from paraquat (PQ) poisoning constitutes a significant mortality risk, predominantly due to oxidative stress and mitochondrial dysfunction. Despite this, effective treatment options are currently limited. PURPOSE This study examines the protective effects of Glabridin (Glab), a flavonoid with noted antioxidant properties, against PQ-induced ALI, with a focus on its mitochondrial function and modulation of oxidative stress. METHODS The research utilized human normal lung epithelial line BEAS-2B cells (B2B) and PQ-exposed C57BL/6J mice to evaluate the role of Glab. Assessments included lung inflammation, oxidative stress markers, and mitochondrial dysfunction, as well as the involvement of the cGAS-STING and caspase-3 pathways. Molecular docking and western blot analyses were used to investigate the interaction between Glab and malic enzyme 1 (ME1). RESULTS The findings indicate that Glab significantly enhances survival rates, reduces inflammation, and mitigates oxidative stress in PQ-exposed mice. In vitro experiments demonstrated that Glab inhibited the cGAS-STING and caspase-3 pathways, release of mitochondrial contents (cytochrome C and mtDNA), decreased mitochondrial ROS production, and stabilized ME1, resulting in increased NADPH levels. CONCLUSION Glab confers protection against PQ-induced ALI by modulating oxidative stress, preserving mitochondrial function, and inhibiting both inflammatory and apoptotic pathways. Notably, the stabilization of malic enzyme 1 (ME1) and the consequent increase in NADPH levels play a critical role in this protective mechanism. These results underscore the potential of Glab as a therapeutic agent for addressing PQ poisoning, with particular emphasis on the pivotal role of ME1 in mediating its effects.
Collapse
Affiliation(s)
- Mingming Zhao
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China; CixiBiomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Xuanhai Xie
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Yitian Ding
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Qiang Zhang
- Emergency Department, The People's Hospital of Yuhuan, Taizhou, 317600, China
| | - Xinheng Liu
- Wenzhou Medical University, Wenzhou, 325000, China
| | - Taotao Lin
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreaic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Guangliang Hong
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
9
|
Hou W, Zhao Y, Yang L, Duan C, Li F, Liu X, Sun W, Gao L. SIRT5-mediated desuccinylation prevents mitochondrial dysfunction in alveolar epithelial cells senescence and pulmonary fibrosis. Cell Signal 2025; 132:111830. [PMID: 40311988 DOI: 10.1016/j.cellsig.2025.111830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/03/2025]
Abstract
Senescence of alveolar epithelial cells (AEC) is a key event in the onset and progression of Idiopathic pulmonary fibrosis (IPF). The pathogenic mechanisms that underlie the effects of AEC senescence remain largely unexplained. Some age-related diseases have an etiology linked to mitochondrial dysfunction induced by excessive lysine succinylation (Ksucc). SIRT5 can remove excessive Ksucc levels to maintain mitochondrial homeostasis. Therefore, this study aimed to determine the effects of SIRT5-mediated de-Ksucc on mitochondrial function and pulmonary fibrosis after AEC senescence. We found AEC in the lungs derived from IPF patients exhibit a marked accumulation of dysmorphic and dysfunctional mitochondria and excessive Ksucc levels. These mitochondrial abnormalities in AEC of normal mice with advancing age were associated with the downregulation of SIRT5. Increased SIRT5 expression by LV-SIRT5pcDNA in senescent AEC sustains mitochondrial integrity and reduces fibrotic effects of AEC senescence in established bleomycin (BLM)-aging mouse model. The level of ITGB1 K238 was upregulation in senescent AEC, LV-SIRT5pcDNA down-regulates the Ksucc level of ITGB1 K238 blocking the activation of ITGB1/STAT3 signaling pathway associated pulmonary fibrosis. Collectively, our findings indicate excessive lysine succinylation (hyperKsucc) is a fundamental basis for mitochondrial dysfunction in pulmonary fibrosis induced by the AEC senescence and SIRT5 alleviates AEC senescence by stabilizing the mitochondrial function.
Collapse
Affiliation(s)
- Wenyu Hou
- The University of Electronic Science and Technology of China, Chengdu 610056, China
| | - Yunmulan Zhao
- The University of Electronic Science and Technology of China, Chengdu 610056, China
| | - Liqing Yang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Chunyan Duan
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Fei Li
- Department of Pulmonary and Critical Care Medicine, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiaoman Liu
- Department of Pulmonary and Critical Care Medicine, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wei Sun
- Department of Pulmonary and Critical Care Medicine, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Lingyun Gao
- Department of Pulmonary and Critical Care Medicine, Second Hospital of Tianjin Medical University, Tianjin 300211, China; Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, Chengdu 610072, China; Department of Pulmonary and Critical Care Medicine, Ziyang People's Hospital, Ziyang 641300, China.
| |
Collapse
|
10
|
Zhang MY, Liu YC, Liu XY, Chen DW, Han C, Shen X, Ding YX, Wang XP, Shi AP. Withaferin A ameliorates psoriasis-like skin lesions by suppressing oxidative stress in keratinocytes. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-13. [PMID: 40279167 DOI: 10.1080/10286020.2025.2492825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 04/07/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025]
Abstract
Psoriasis is a chronic disease with elusive pathogenesis linked to genetic, and immune factors. Studies have shown most patients have experienced high levels of oxidative stress, which can lead to inflammation or damage. Withaferin A is a natural product with multiple pharmacological activities, including anti-tumor and anti-inflammatory effects. We investigated the effects of WA in treating Imiquimod (IMQ)-induced psoriasis mice. The epidermal pathology of mice was significantly improved after treatment. WA inhibited inflammation by decreasing the production of IL-1β, IL-6, and IFN-γ, which were induced by epidermal oxidative stress. Additionally, WA can shift the macrophage status from pro-inflammatory to anti-inflammatory.
Collapse
Affiliation(s)
- Ming-Yi Zhang
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yu-Chang Liu
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xing-Yu Liu
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Di-Wei Chen
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Han
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xin Shen
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - You-Xiang Ding
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xiao-Ping Wang
- Department of Basic Medicine, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ai-Ping Shi
- Department of Pharmacy, Taixing People's Hospital, Taixing 225400, China
| |
Collapse
|
11
|
Zhang F, Wang T, Wei L, Xie Z, Wang L, Luo H, Li F, Kang Q, Dong W, Zhang J, Zhu X, Wang C, Liang L, Peng W. B-Lymphoid Tyrosine Kinase Crosslinks Redox and Apoptosis Signaling Networks to Promote the Survival of Transplanted Bone Marrow Mesenchymal Stem Cells. RESEARCH (WASHINGTON, D.C.) 2025; 8:0660. [PMID: 40235595 PMCID: PMC11999575 DOI: 10.34133/research.0660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 02/25/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
Stress-induced apoptosis presents an obstacle to bone marrow mesenchymal stem cell (BMSC) transplantation to repair steroid-induced osteonecrosis of the femoral head (SONFH). Thus, appropriate intervention strategies should be explored to mitigate this. In our previous study, we discovered a new subgroup of BMSCs-the oxidative stress-resistant BMSCs (OSR-BMSCs)-which can survive the oxidative stress microenvironment in the osteonecrotic area, through a mechanism that currently remains unclear. In this study, we found that B-lymphoid tyrosine kinase (BLK) may be the crucial factor regulating the oxidative stress resistance of OSR-BMSCs, as it is highly expressed in these cells. Knockdown of BLK eliminated oxidative stress resistance, aggravated oxidative stress-induced apoptosis, reduced the survival of OSR-BMSCs in the oxidative stress microenvironment of the osteonecrotic area, and greatly weakened the transplantation efficacy of OSR-BMSCs for SONFH. By contrast, BLK was weakly expressed in oxidative stress-sensitive BMSCs (OSS-BMSCs). Overexpression of BLK in susceptible OSS-BMSCs allowed them to acquire oxidative stress resistance, inhibited oxidative stress-induced apoptosis, promoted their survival in the osteonecrotic area, and improved the transplantation efficacy of OSS-BMSCs for SONFH. Mechanistically, BLK concurrently activates redox and apoptotic signaling networks through its tyrosine kinase activity, which confers oxidative stress resistance to BMSCs and inhibits their stress-induced apoptosis of BMSCs. Herein, we report that OSR-BMSCs have intrinsic oxidative stress resistance that is conferred and mediated by BLK. This finding provides a potential new intervention strategy for improving the survival of transplanted BMSCs and the therapeutic efficacy of BMSC transplantation for SONFH.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Tao Wang
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Lei Wei
- Department of Orthopedics,
Warren Alpert Medical School of Brown University,Providence, RI 02912, USA
| | - Zhihong Xie
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Lijun Wang
- Department of Critical Care Medicine,
West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Hong Luo
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Fanchao Li
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Qinglin Kang
- Department of Orthopedics,
Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Wentao Dong
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Jian Zhang
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Xuesong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Chuan Wang
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Liang Liang
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| | - Wuxun Peng
- Department of Emergency Surgery,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
- Laboratory of Emergency Medicine,
The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| |
Collapse
|
12
|
Zhang Z, Luo Y, Zhang H, Zeng Z, Zheng W, Zhao Y, Huang Y, Shen L. Exploring the mechanisms of cow placental peptides in delaying liver aging based on mitochondrial energy metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119593. [PMID: 40064320 DOI: 10.1016/j.jep.2025.119593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Placenta is a kind of traditional Chinese medicine, known as "Ziheche". The role of cow placental peptides (CPP) in delaying liver aging has been reported, and in-depth exploration of the specific regulatory mechanisms is of great significance for the recycling and utilization of CPP and the development of natural anti-aging drugs. AIM OF THE STUDY To investigate the protective effects and mechanisms of CPP on liver aging induced by D-galactose (D-gal) in mice from the perspective of mitochondrial energy metabolism. METHODS An aging model was induced in mice using D-gal. The body weight and liver index of mice were measured, followed by staining and electron microscopy to observe liver morphology and aging markers. Reactive oxygen species (ROS) levels and antioxidant-related indicators were assessed, and mitochondrial function was evaluated. Finally, changes and mechanisms in liver transcriptomics and targeted mitochondrial energy metabolomics were analyzed and integrated to elucidate the regulatory pathways through which CPP delays liver aging. RESULTS CPP improved liver structural damage, oxidative stress, and mitochondrial dysfunction induced by D-galactose in aging mice. It increased the final body weight and liver index, alleviated hepatocyte swelling and degeneration, enhanced liver antioxidant capacity, and restored normal mitochondrial morphology and function. The combined analysis of targeted mitochondrial energy metabolomics and liver transcriptomics revealed that CPP directly or indirectly regulated mitochondrial energy metabolism and delayed aging by influencing the cAMP signaling pathway, PI3K-Akt signaling pathway, oxidative phosphorylation, and other pathways, thereby modulating related genes and metabolites.
Collapse
Affiliation(s)
- Zeru Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuxin Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hanwen Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Zhi Zeng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Weijian Zheng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuquan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yixin Huang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
13
|
Elmorsy EM, Al-Ghafari AB, Al Doghaither HA, Alrowaili MG, Khired ZA, Toraih EA, Fawzy MS, Shehata SA. Vitamin D Alleviates Heavy Metal-Induced Cytotoxic Effects on Human Bone Osteoblasts Via the Induction of Bioenergetic Disruption, Oxidative Stress, and Apoptosis. Biol Trace Elem Res 2025; 203:2420-2434. [PMID: 39235540 PMCID: PMC11920318 DOI: 10.1007/s12011-024-04337-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/02/2024] [Indexed: 09/06/2024]
Abstract
Cadmium (Cd) and lead (Pb) are heavy metals (HMs) that persistently contaminate the ecosystem, and bioaccumulation in bones is a health concern. We used biochemical and molecular assays to assess the cytoprotective effect of vitamin D (VD) on Cd- and Pd-induced chemical toxicity of human bone osteoblasts in vitro. Exposing Cd and Pb to human osteoblast cultures at concentrations of 0.1-1000 µM for 24-72 h significantly reduced osteoblast viability in an exposure time- and concentration-dependent manner. The cytotoxic effect of Cd on osteoblasts was more severe than Pb's, with 72-h exposure estimated half maximal effective concentration (EC50) of 8 and 12 µM, respectively, and VD (1 and 10 nM) alleviated cytotoxicity. Bioenergetics assays of ATP, mitochondrial membrane potential, and mitochondrial complex I and III activity showed that both Cd and Pb (1 and 10 µM) inhibited cellular bioenergetics after 72-h exposure. Cd and Pb increased lipid peroxidation and reactive oxygen species with reduced catalase/superoxide dismutase antioxidant activities and increased activity of caspases -3, -8, and -9. Co-treatment with VD (1 and 10 nM) counteracted bioenergetic disruption, oxidative damage, and apoptosis in a concentration-dependent manner. These findings suggest that VD is effective in managing the toxic effects of environmental pollutants and in treating bone diseases characterized by oxidative stress, apoptosis, and bioenergetic disruption.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Pathology Department, Faculty of Medicine, Northern Border University, 91431, Arar, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Majed Gorayan Alrowaili
- Department of Surgery (Orthopedic Division), Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| | - Zenat Ahmed Khired
- Department of Surgery, College of Medicine, Jazan University, 45142, Jazan, Saudi Arabia
| | - Eman A Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
- Genetics Unit, Department of Histology and Cell Biology, Suez Canal University, Ismailia, 41522, Egypt.
| | - Manal S Fawzy
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
- Department of Biochemistry, Faculty of Medicine, Northern Border University, 73213, Arar, Saudi Arabia
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Shaimaa A Shehata
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
14
|
Ailioaie LM, Ailioaie C, Litscher G. Fighting Cancer with Photodynamic Therapy and Nanotechnologies: Current Challenges and Future Directions. Int J Mol Sci 2025; 26:2969. [PMID: 40243613 PMCID: PMC11989081 DOI: 10.3390/ijms26072969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Photodynamic therapy (PDT) is an innovative treatment that has recently been approved for clinical use and holds promise for cancer patients. It offers several benefits, such as low systemic toxicity, minimal invasiveness, and the ability to stimulate antitumor immune responses. For certain types of cancer, it has shown positive results with few side effects. However, PDT still faces some challenges, including limited light penetration into deeper tumor tissues, uneven distribution of the photosensitizer (PS) that can also affect healthy cells, and the difficulties posed by the hypoxic tumor microenvironment (TME). In hypoxic conditions, PDT's effectiveness is reduced due to insufficient production of reactive oxygen species, which limits tumor destruction and can lead to relapse. This review highlights recent advances in photosensitizers and nanotechnologies that are being developed to improve PDT. It focuses on multifunctional nanoplatforms and nanoshuttles that have shown promise in preclinical studies, especially for treating solid tumors. One of the key areas of focus is the development of PSs that specifically target mitochondria to treat deep-seated malignant tumors. New mitochondria-targeting nano-PSs are designed with better water solubility and extended wavelength ranges, allowing them to target tumors more effectively, even in challenging, hypoxic environments. These advancements in PDT are opening new doors for cancer treatment, especially when combined with other therapeutic strategies. Moving forward, research should focus on optimizing PDT, creating more efficient drug delivery systems, and developing smarter PDT platforms. Ultimately, these efforts aim to make PDT a first-choice treatment option for cancer patients.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania; (L.M.A.); (C.A.)
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania; (L.M.A.); (C.A.)
| | - Gerhard Litscher
- Swiss University of Traditional Chinese Medicine, SWISS TCM UNI, High-Tech Acupuncture and Digital Chinese Medicine, 5330 Bad Zurzach, Switzerland
- President of the International Society for Medical Laser Applications (ISLA Transcontinental), German Vice President of the German-Chinese Research Foundation (DCFG) for TCM, Honorary President of the European Federation of Acupuncture and Moxibustion Societies, Honorary Professor of China Beijing International Acupuncture Training Center, China Academy of Chinese Medical Sciences, Honorary President of the American Association of Laser Acupuncture Therapy (ASLAT), USA, Former Head of Two Research Units and the TCM Research Center at the Medical University of Graz, 8053 Graz, Austria
| |
Collapse
|
15
|
Han L, Ho CT, Lu M. Regulatory Role of Bioactive Compounds from Natural Spices on Mitochondrial Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5711-5723. [PMID: 40019340 DOI: 10.1021/acs.jafc.4c12341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Natural spices have gained much attention for their aromatic and pungent flavors as well as their multiple beneficial health effects. As complex organelles that play a central role in energy production, stress response control, cell signal transduction, and metabolism regulation, mitochondria could be regulated by many bioactive components in spices. In this review, the role of mitochondria in maintaining cellular and metabolism homeostasis is summarized. The regulatory effects of mitochondrial function by major bioactive compounds from natural spices are evaluated, including capsaicin, 6-gingerol, 6-shogaol, allicin, quercetin, curcumin, tetrahydrocurcumin, and cinnamaldehyde. The underlying molecular mechanisms are also discussed. This work could enhance our understanding toward health-promoting properties of spice compounds as well as provide new insights into the prevention and treatment of disorders associated with mitochondrial dysfunctions by those nutraceuticals.
Collapse
Affiliation(s)
- Liguang Han
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
16
|
Feng W, Chen N, Chen K, Chen T. Forkhead box O-1 regulates the biological behavior of BMP-2-induced human bone mesenchymal stem cells through mitochondrial dynamics and autophagy. BIOMOLECULES & BIOMEDICINE 2025; 25:869-882. [PMID: 39226107 PMCID: PMC11959391 DOI: 10.17305/bb.2024.10686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 09/05/2024]
Abstract
This study explored the mechanism by which forkhead box O-1 (FoxO1) modulates the biological behaviors of bone mesenchymal stem cell (BMSC). Human BMSCs were cultured for seven days in Dulbecco's modified Eagle medium (DMEM) containing bone morphogenetic protein-2 (BMP-2) and treated with a short hairpin-FoxO1 plasmid. The study assessed cell proliferation, migration, apoptosis, adenosine triphosphate (ATP) levels, mitochondrial DNA (mtDNA) levels, membrane potential (MMP), autophagy, and the levels of FoxO1, apoptosis-associated proteins, osteogenic differentiation-associated proteins, mitochondrial fusion and fission proteins, and mitochondrial autophagy-related proteins. The cells were also treated with the mitochondrial fusion activator MASM7 and the mitochondrial autophagy activator carbonyl cyanide 3-chlorophenylhydrazone (CCCP). The study evaluated whether mitochondrial dynamics and autophagy activation could rescue the FoxO1 knockdown-induced changes in BMSC biological behaviors, mitochondrial dynamics, and mitochondrial autophagy. BMP-2-induced BMSCs exhibited upregulated FoxO1 expression, enhanced proliferation and migration, and induced osteogenic differentiation, while FoxO1 knockdown inhibited BMP-2-induced BMSC proliferation, migration and osteogenic differentiation, increased apoptosis, and affected mitochondrial dynamics and autophagy. Promoting mitochondrial fusion partially reversed the regulatory effects of FoxO1 downregulation on mitochondrial autophagy and the inhibitory effects of FoxO1 silencing on BMP-2-induced BMSC biological behaviors. Activated mitochondrial autophagy facilitated the homeostasis of mitochondrial dynamics and partially counteracted the inhibitory effects of FoxO1 knockdown on BMP-2-induced BMSC biological behaviors. In conclusion, FoxO1 regulates mitochondrial dynamics and autophagy to modulate the osteogenic differentiation of BMP-2-induced human BMSCs.
Collapse
Affiliation(s)
- Weijia Feng
- Department of Pediatric Orthopaedic, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Nannan Chen
- The School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Chen
- Shanghai Key Laboratory of Materials Laser Processing and Modification, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Chen
- Department of Pediatric Orthopaedic, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Marciniak E, Osuch B, Młotkowska P, Kowalczyk P, Roszkowicz-Ostrowska K, Misztal T. Gene Expression and Activity of Selected Antioxidant and DNA Repair Enzymes in the Prefrontal Cortex of Sheep as Affected by Kynurenic Acid. Int J Mol Sci 2025; 26:2381. [PMID: 40141025 PMCID: PMC11942221 DOI: 10.3390/ijms26062381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
The prefrontal cortex (PCx) is involved in many higher-order cognitive processes, including decision making, reasoning, personality expression, and social cognition. These functions are associated with high energy demand and the production of harmful oxygen radicals. Recent studies indicate that kynurenic acid (KYNA) exerts neuroprotective effects, largely due to its anti-inflammatory and antioxidant properties. To further evaluate the antioxidant potential of this compound, we tested the hypothesis that increasing KYNA levels in the sheep cerebroventricular circulation would positively affect the mRNA expression and activity of selected antioxidant and DNA repair enzymes in the distal part of the brain, i.e., the PCx. Anestrous sheep were infused intracerebroventricularly with a series of two KYNA doses: lower (4 × 5 μg/60 μL/30 min) and higher (4 × 25 μg/60 μL/30 min) at 30 min intervals. The results demonstrated that KYNA exerted significant dose-dependent stimulatory effects on the activity of superoxide dismutase 2, catalase, and glutathione peroxidase 1 while inhibiting their transcription in a similar manner. In addition, KYNA was also found to dose-dependently activate the base excision repair pathway, as determined by the increased transcript levels of glycosylases: N-methylpurine DNA glycosylase, thymine-DNA glycosylase, 8-oxoguanine DNA glycosylase-1, and apurinic/apyrimidinic endonuclease 1. The excision efficiency of damaged nucleobases, such as εA, εC and 8-oxoG, by these enzymes was also increased in response to central KYNA infusion. These findings expand the knowledge on KYNA as a potential protective factor against oxidative stress in the central nervous system.
Collapse
Affiliation(s)
| | | | | | | | | | - Tomasz Misztal
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3 Street, 05-110 Jabłonna, Poland; (E.M.); (B.O.); (P.M.); (P.K.); (K.R.-O.)
| |
Collapse
|
18
|
Zhang J, Chen Z, Wang S, Kong L, Liu J. Prodelphinidin from purple sweet potato induces apoptosis in human triple-negative breast cancer cells via ROS-mediated ER stress activation. Med Oncol 2025; 42:92. [PMID: 40048036 DOI: 10.1007/s12032-025-02642-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Triple negative breast cancer (TNBC), a highly aggressive and heterogeneous subtype of breast cancer, lacks an effective targeted therapy. Conventional medication has limited efficacy in treating TNBC, which highlights the potential of developing therapeutic agents from natural bioactive compounds. This study aimed to investigate the cytotoxicity of prodelphinidin (PD), an anthocyanin found in purple sweet potato, in human MDA-MB-231 and MDA-MB-436 cells. The results showed that PD selectively inhibited human breast cancer, particularly TNBC. Furthermore, PD demonstrated significant dose- and time-dependent inhibition of MDA-MB-231 and MDA-MB-436 cell activity. Flow cytometry and western blot analysis revealed that PD induced cell apoptosis by down-regulating Bcl-2, activating caspase-3/9, and cleaving PARP. Additionally, PD treatment upregulated the expression of p-elF2α, GRP78, and CHOP, indicating the involvement of endoplasmic reticulum stress (ERS). PD treatment also increased the production of reactive oxygen species (ROS) and decreased superoxide dismutase (SOD) activity in TNBC cells. The cytotoxicity of PD reduced significantly by pre-treatment with caspase inhibitors (Ac-DEVD-CHO and Z-LEHD-FMK). In conclusion, PD effectively inhibited the proliferation and induced apoptosis in TNBC cells through the activation of ROS and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Jiluan Zhang
- Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China
| | - Zihan Chen
- Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China
| | - Shibo Wang
- Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China
| | - Lingxue Kong
- Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China
| | - Jinjuan Liu
- Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, 221116, China.
| |
Collapse
|
19
|
de Oliveira MP, da Silva LE, Fernandes BB, Steiner MR, Pistóia DG, Santos Cichella TD, Jacinto LB, Spuldaro KM, Pinto Moehlecke Iser B, Rezin GT. The impact of obesity on mitochondrial dysfunction during pregnancy. Mol Cell Endocrinol 2025; 598:112463. [PMID: 39832615 DOI: 10.1016/j.mce.2025.112463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Mitochondria play a central role in nutrient metabolism, besides being responsible for the production of adenosine triphosphate (ATP), the main source of cellular energy. However, the ATP production process is associated with the generation of reactive oxygen species (ROS), which excessive accumulation can cause mitochondrial dysfunction. This dysfunction, in turn, causes the accumulation of fatty acids in the adipose tissue, triggering a local inflammatory process that can evolve into systemic inflammation. In women with obesity, an increase in lipid levels in the placental environment is observed. The high presence of fatty acids compromises the structural integrity and mitochondrial membrane, culminating in the release of ROS. This process damages the DNA of placental cells and causes an inflammatory state, affecting metabolic efficiency. This vicious cycle is characterized by defects in mitochondrial ATP production, which can lead to lipid accumulation and inflammation. In pregnant women with obesity, these mitochondrial changes play a determining role in pregnancy outcomes. Hence, the objective of this study was to search the literature to review the impact of mitochondrial dysfunction in the maternal obesity.
Collapse
Affiliation(s)
- Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil.
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Bruna Barros Fernandes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Mariella Reinol Steiner
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Debora Gehrke Pistóia
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Tamires Dos Santos Cichella
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Luana Bahia Jacinto
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Karoline Marcondes Spuldaro
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Betine Pinto Moehlecke Iser
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Tubarão, Santa Catarina, Brazil
| |
Collapse
|
20
|
Ye X, Yang R, Riaz T, Chen J. Stability and antioxidant function of Porphyra haitanensis proteins during simulated gastrointestinal digestion: Effects on stress resistance and lifespan extension in Caenorhabditis elegans. Int J Biol Macromol 2025; 293:139291. [PMID: 39740706 DOI: 10.1016/j.ijbiomac.2024.139291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
Porphyra haitanensis proteins (PHP) are natural proteins with various nutritional and biological values. This study was to analyze the composition, stability, and antioxidant activity of PHP before and after simulation gastrointestinal digestion (SGD). Caenorhabditis elegans was used as the model to investigate the functional activity and potential mechanisms of action of the PHP digestion products (PHPDP). The results showed that PHP contained 16 amino acids and exhibited high thermal stability (up to 80 °C), but dimerization or fragmentation occurred in environments with pH 3 and pH 11. After digestion, PHP released 212 bioactive peptides, which significantly enhanced its antioxidant activity and improved the resistance of C. elegans to heat, oxidative, and ultraviolet stress. Furthermore, PHPDP improved oxidative stress in C. elegans and extended its lifespan by increasing antioxidant enzyme activity and reducing malondialdehyde, lipofuscin, and reactive oxygen species levels. The mechanism of action of PHPDP likely involved regulating the insulin signaling pathway through daf-2/daf-16 and modulating the expression of oxidative stress regulators skn-1 and sod-3, thereby enhancing the organism's stress resistance and extending lifespan. This study demonstrated that P. haitanensis could serve as a reliable protein source in daily life and provided a reference for its development and application as a functional food ingredient.
Collapse
Affiliation(s)
- Xianjiang Ye
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; National Research and Development Centre for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ran Yang
- National Research and Development Centre for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Talha Riaz
- National Research and Development Centre for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jicheng Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| |
Collapse
|
21
|
Zeng JQ, Zhou HF, Du HX, Wu YJ, Mao QP, Yin JJ, Wan HT, Yang JH. Tongmai Hypoglycemic Capsule Attenuates Myocardial Oxidative Stress and Fibrosis in the Development of Diabetic Cardiomyopathy in Rats. Chin J Integr Med 2025; 31:251-260. [PMID: 39644459 DOI: 10.1007/s11655-024-4002-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE To investigate the effect of Tongmai Hypoglycemic Capsule (THC) on myocardium injury in diabetic cardiomyopathy (DCM) rats. METHODS A total of 24 Sprague Dawley rats were fed for 4 weeks with high-fat and high-sugar food and then injected with streptozotocin intraperitoneally for the establishment of the DCM model. In addition, 6 rats with normal diets were used as the control group. After modeling, 24 DCM rats were randomly divided into the model, L-THC, M-THC, and H-THC groups by computer generated random numbers, and 0, 0.16, 0.32, 0.64 g/kg of THC were adopted respectively by gavage, with 6 rats in each group. After 12 weeks of THC administration, echocardiography, histopathological staining, biochemical analysis, and Western blot were used to detect the changes in myocardial structure, oxidative stress (OS), biochemical indexes, protein expressions of myocardial fibrosis, and nuclear factor erythroid 2-related faactor 2 (Nrf2) element, respectively. RESULTS Treatment with THC significantly decreased cardiac markers such as creatine kinase, lactate dehydrogenase, and creatine kinase-MB, etc., (P<0.01); enhanced cardiac function indicators including heart rate, ejection fraction, cardiac output, interventricular septal thickness at diastole, and others (P<0.05 or P<0.01); decreased levels of biochemical indicators such as fasting blood glucose, total cholesterol, triglycerides, low-density lipoprotein cholesterol, aspartate transaminase, (P<0.05 or P<0.01); and decreased the levels of myocardial fibrosis markers α-smooth muscle actin (α-SMA), and collagen I (Col-1) protein (P<0.01), improved myocardial morphology and the status of myocardial interstitial fibrosis. THC significantly reduced malondialdehyde levels in model rats (P<0.01), increased levels of catalase, superoxide dismutase, and glutathione (P<0.01), and significantly increased the expression of Nrf2, NAD(P)H:quinone oxidoreductase 1, heme oxygenase-1, and superoxide dismutase 2 proteins in the left ventricle of rats (P<0.01). CONCLUSION THC activates the Nrf2 signaling pathway and plays a protective role in reducing OS injury and cardiac fibrosis in DCM rats.
Collapse
Affiliation(s)
- Jie-Qiong Zeng
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Hui-Fen Zhou
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Hai-Xia Du
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Yu-Jia Wu
- College of Life Science, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Qian-Ping Mao
- College of Life Science, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Jun-Jun Yin
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Hai-Tong Wan
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China
| | - Jie-Hong Yang
- College of Basic Medicine Sciences, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310051, China.
| |
Collapse
|
22
|
Naiki‐Ito A, Naiki T, Takahashi S. Exploring experimental models of prostate cancer in chemoprevention: Oxidative stress as a key pathway to translational research. Pathol Int 2025; 75:131-144. [PMID: 39807695 PMCID: PMC11922031 DOI: 10.1111/pin.13509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Prostate cancer (PCa) is the second most common cancer in men globally. Its growth is driven by oxidative stress associated with inflammation, aging, and environmental factors, including diet and lifestyle. These factors contribute to multiple stages of PCa progression, including progression to castration-resistant prostate cancer (CRPC). Therefore, oxidative stress represents an intriguing target for PCa chemoprevention and treatment. In vivo experimental models are crucial for understanding the mechanisms of PCa development, validating chemopreventive and therapeutic approaches, and translating preclinical results into clinical applications. We established a transgenic rat for adenocarcinoma of the prostate (TRAP) model, a transgenic rat that efficiently develops androgen-dependent adenocarcinoma, pathologically and biologically mimicking human PCa progression, to clarify the mechanisms of tumor progression, including the involvement of oxidative stress, and established a system for screening the chemopreventive effects of agents against PCa. Additionally, we derived a CRPC model from the TRAP model and developed a distant metastasis model, providing a comprehensive multistage rat model of prostate carcinogenesis. This review presents findings on the molecular mechanisms of PCa and the chemopreventive effects of natural compounds with antioxidant properties, such as polyphenols. We additionally described the potential for repositioning existing drugs with antiandrogenic activity for PCa chemoprevention.
Collapse
Affiliation(s)
- Aya Naiki‐Ito
- Department of Experimental Pathology and Tumor BiologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Taku Naiki
- Department of Experimental Pathology and Tumor BiologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
- Department of Nephro‐urologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor BiologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| |
Collapse
|
23
|
Wang H, Wang Z, Wu Q, Yang Y, Liu S, Bian J, Bo L. Perioperative oxygen administration for adults undergoing major noncardiac surgery: a narrative review. Med Gas Res 2025; 15:73-84. [PMID: 39436170 PMCID: PMC11515063 DOI: 10.4103/mgr.medgasres-d-24-00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/29/2024] [Accepted: 04/07/2024] [Indexed: 10/23/2024] Open
Abstract
Perioperative oxygen administration, a topic under continuous research and debate in anesthesiology, strives to optimize tissue oxygenation while minimizing the risks associated with hyperoxia and hypoxia. This review provides a thorough overview of the current evidence on the application of perioperative oxygen in adult patients undergoing major noncardiac surgery. The review begins by describing the physiological reasoning for supplemental oxygen during the perioperative period and its potential benefits while also focusing on potential hyperoxia risks. This review critically appraises the existing literature on perioperative oxygen administration, encompassing recent clinical trials and meta-analyses, to elucidate its effect on postoperative results. Future research should concentrate on illuminating the optimal oxygen administration strategies to improve patient outcomes and fine-tune perioperative care protocols for adults undergoing major noncardiac surgery. By compiling and analyzing available evidence, this review aims to provide clinicians and researchers with comprehensive knowledge on the role of perioperative oxygen administration in major noncardiac surgery, ultimately guiding clinical practice and future research endeavors.
Collapse
Affiliation(s)
- Huixian Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qi Wu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yuguang Yang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shanshan Liu
- Department of Anesthesiology, Chenggong Hospital Affiliated to Xiamen University, Xiamen, Fujian Province, China
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lulong Bo
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Cabrera-Serrano AJ, Sánchez-Maldonado JM, González-Olmedo C, Carretero-Fernández M, Díaz-Beltrán L, Gutiérrez-Bautista JF, García-Verdejo FJ, Gálvez-Montosa F, López-López JA, García-Martín P, Pérez EM, Sánchez-Rovira P, Reyes-Zurita FJ, Sainz J. Crosstalk Between Autophagy and Oxidative Stress in Hematological Malignancies: Mechanisms, Implications, and Therapeutic Potential. Antioxidants (Basel) 2025; 14:264. [PMID: 40227235 PMCID: PMC11939785 DOI: 10.3390/antiox14030264] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy is a fundamental cellular process that maintains homeostasis by degrading damaged components and regulating stress responses. It plays a crucial role in cancer biology, including tumor progression, metastasis, and therapeutic resistance. Oxidative stress, similarly, is key to maintaining cellular balance by regulating oxidants and antioxidants, with its disruption leading to molecular damage. The interplay between autophagy and oxidative stress is particularly significant, as reactive oxygen species (ROS) act as both inducers and by-products of autophagy. While autophagy can function as a tumor suppressor in early cancer stages, it often shifts to a pro-tumorigenic role in advanced disease, aiding cancer cell survival under adverse conditions such as hypoxia and nutrient deprivation. This dual role is mediated by several signaling pathways, including PI3K/AKT/mTOR, AMPK, and HIF-1α, which coordinate the balance between autophagic activity and ROS production. In this review, we explore the mechanisms by which autophagy and oxidative stress interact across different hematological malignancies. We discuss how oxidative stress triggers autophagy, creating a feedback loop that promotes tumor survival, and how autophagic dysregulation leads to increased ROS accumulation, exacerbating tumorigenesis. We also examine the therapeutic implications of targeting the autophagy-oxidative stress axis in cancer. Current strategies involve modulating autophagy through specific inhibitors, enhancing ROS levels with pro-oxidant compounds, and combining these approaches with conventional therapies to overcome drug resistance. Understanding the complex relationship between autophagy and oxidative stress provides critical insights into novel therapeutic strategies aimed at improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Carmen González-Olmedo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - María Carretero-Fernández
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - Leticia Díaz-Beltrán
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Juan Francisco Gutiérrez-Bautista
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Servicio de Análisis Clínicos e Inmunología, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Department of Biochemistry, Molecular Biology and Immunology III, University of Granada, 18016 Granada, Spain
| | - Francisco José García-Verdejo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Gálvez-Montosa
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - José Antonio López-López
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Paloma García-Martín
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Eva María Pérez
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Pedro Sánchez-Rovira
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Jesús Reyes-Zurita
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
25
|
Chen L, Tang J, Zuo X, Li B, Liu C, Hong S, Min J, Hu M, Li S, Zhou M, Chen M, He Y, Xiao Y, Huang X, Hong L. SIRT1 Alleviates Oxidative Stress-Induced Mitochondrial Dysfunction and Mitochondria-Associated Membrane Dysregulation in Stress Urinary Incontinence. Cell Prolif 2025:e70009. [PMID: 39980436 DOI: 10.1111/cpr.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/21/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025] Open
Abstract
The pathogenesis of stress urinary incontinence (SUI), a condition common in women, remains to be fully elucidated. This study revealed that the incidence of SUI is associated with mitochondrial homeostasis dysregulation following oxidative stress in the fibrous connective tissue of the pelvic floor. SIRT1 is an essential factor for maintaining mitochondrial homeostasis; however, its potential role and mechanism of action in SUI pathogenesis remain unclear. Both in vitro and in vivo, we observed that oxidative stress reduced SIRT1 expression to inhibit the PGC-1α/NRF1/TFAM and PINK1/Parkin signalling pathways, eliciting impairment of mitochondrial biogenesis and mitophagy in L929 cells and SUI mice. Decreased SIRT1 levels induced endoplasmic reticulum (ER) stress and altered the structure of mitochondria-associated membranes (MAMs), disrupting ER-mitochondrial calcium homeostasis and exacerbting ROS accumulation. SIRT1 activation can restore mitochondrial function and the structure of MAMs and alleviate ER stress in fibroblasts, promoting anterior vaginal wall repair and improving urodynamic parameters in the SUI model. Our findings provide novel insights into the role and associated mechanism of SIRT1 in ameliorating oxidative stress-induced mitochondrial dysfunction in fibroblasts of the anterior vaginal wall and propose SIRT1 as a potential therapeutic target for SUI.
Collapse
Affiliation(s)
- Liying Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Jianming Tang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Xiaohu Zuo
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Bingshu Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Cheng Liu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Shasha Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Jie Min
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Ming Hu
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Suting Li
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Min Zhou
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Mao Chen
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yong He
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Ya Xiao
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Xiaoyu Huang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Li Hong
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
26
|
Yu X, Gao J, Zhang C. Sepsis-induced cardiac dysfunction: mitochondria and energy metabolism. Intensive Care Med Exp 2025; 13:20. [PMID: 39966268 PMCID: PMC11836259 DOI: 10.1186/s40635-025-00728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Sepsis is a life-threatening multi-organ dysfunction syndrome caused by dysregulated host response to infection, posing a significant global healthcare challenge. Sepsis-induced myocardial dysfunction (SIMD) is a common complication of sepsis, significantly increasing mortality due to its high energy demands and low compensatory reserves. The substantial mitochondrial damage rather than cell apoptosis in SIMD suggests disrupted cardiac energy metabolism as a crucial pathophysiological mechanism. Therefore, we systematically reviewed the mechanisms underlying energy metabolism dysfunction in SIMD, including alterations in myocardial cell energy metabolism substrates, excitation-contraction coupling processes, mitochondrial dysfunction, and mitochondrial autophagy and biogenesis, summarizing potential therapeutic targets within them.
Collapse
Affiliation(s)
- Xueting Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Jie Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxiang Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China.
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China.
- FACC, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
27
|
Yang HM. Mitochondrial Dysfunction in Neurodegenerative Diseases. Cells 2025; 14:276. [PMID: 39996748 PMCID: PMC11853439 DOI: 10.3390/cells14040276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Mitochondrial dysfunction represents a pivotal characteristic of numerous neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. These conditions, distinguished by unique clinical and pathological features, exhibit shared pathways leading to neuronal damage, all of which are closely associated with mitochondrial dysfunction. The high metabolic requirements of neurons make even minor mitochondrial deficiencies highly impactful, driving oxidative stress, energy deficits, and aberrant protein processing. Growing evidence from genetic, biochemical, and cellular investigations associates impaired electron transport chain activity and disrupted quality-control mechanisms, such as mitophagy, with the initial phases of disease progression. Furthermore, the overproduction of reactive oxygen species and persistent neuroinflammation can establish feedforward cycles that exacerbate neuronal deterioration. Recent clinical research has increasingly focused on interventions aimed at enhancing mitochondrial resilience-through antioxidants, small molecules that modulate the balance of mitochondrial fusion and fission, or gene-based therapeutic strategies. Concurrently, initiatives to identify dependable mitochondrial biomarkers seek to detect pathological changes prior to the manifestation of overt symptoms. By integrating the current body of knowledge, this review emphasizes the critical role of preserving mitochondrial homeostasis as a viable therapeutic approach. It also addresses the complexities of translating these findings into clinical practice and underscores the potential of innovative strategies designed to delay or potentially halt neurodegenerative processes.
Collapse
Affiliation(s)
- Han-Mo Yang
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| |
Collapse
|
28
|
Li Q, Wang Z, Jiang C, Yin J, Liu Y, Qu X, Yi X, Gao C. Integration of Transcriptomics and Proteomics to Elucidate Inhibitory Effect and Mechanism of Antifungalmycin B from Marine Streptomyces hiroshimensis in Treating Talaromyces marneffei. Mar Drugs 2025; 23:76. [PMID: 39997200 PMCID: PMC11857274 DOI: 10.3390/md23020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/26/2025] Open
Abstract
Talaromyces marneffei (TM) is an opportunistic pathogenic fungus that mainly infects immunocompromised patients. Currently, the global prevalence of talaromycosis caused by TM is increasing, leading to an increased demand for anti-TM drugs. In our previous study, a novel 28-membered macrolide compound, antifungalmycin B (ANB), was isolated from Streptomyces hiroshimensis GXIMD 06359, exhibiting significant antifungal properties. However, its in vivo mechanisms and direct antifungal effects warrant further investigation. In this study, we employed a mouse model in conjunction with transcriptomic and proteomic approaches to explore the antifungal activity of ANB against T. marneffei. In an in vivo mouse model infected with T. marneffei infection, ANB significantly reduced fungal burdens in the liver, spleen, lungs, and kidneys. Additionally, it markedly decreased the levels of reactive oxygen species (ROS) and cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. Proteomic and transcriptomic studies, complemented by parallel reaction monitoring (PRM) analysis, revealed that ANB effectively disrupted acid biosynthesis and cellular energy metabolism, thereby impairing mitochondrial functions in T. marneffei. These effects were exerted through multiple pathways. These findings highlight the potential of ANB as a versatile inhibitor of polyene macrolide-resistant fungi, offering a promising therapeutic avenue for the treatment of talaromycosis.
Collapse
Affiliation(s)
- Qiqi Li
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Zhou Wang
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Cuiping Jiang
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Jianglin Yin
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Yonghong Liu
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xinjian Qu
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xiangxi Yi
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Chenghai Gao
- Institute of Marine Drugs/Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China; (Q.L.); (Z.W.); (C.J.); (J.Y.); (Y.L.)
- Guangxi Key Laboratory of Marine Drugs/Guangxi University Engineering Research Center of High-Efficient Utilization of Marine Traditional Chinese Medicine Resources, Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
29
|
Wu LL, Shi WD, Peng WF, Li GY. Unraveling the interplay between meningitis and mitochondria: Etiology, pathogenesis, and therapeutic insights. Int Immunopharmacol 2025; 147:113985. [PMID: 39765004 DOI: 10.1016/j.intimp.2024.113985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/11/2024] [Accepted: 12/28/2024] [Indexed: 01/29/2025]
Abstract
Meningitis, characterized by an inflammatory response affecting the membranes surrounding the brain and spinal cord, poses a formidable challenge to global public health. Its etiology spans a spectrum of infectious agents, ranging from bacteria, to viruses, fungi, and parasites. Concurrently, mitochondria-traditionally known as 'cellular powerhouses'-have emerged as critical players in various essential biological functions, including but not limited to, energy production, metabolic regulation, and cell fate determination. Emerging evidence suggests that mitochondria may play vital roles in the pathogenesis of meningitis. In this review, we delineated the definition, classification, etiology, pathogenesis, and clinical manifestations of meningitis, and elucidated the structure, dynamics and functions of mitochondria. We subsequently delved into the intricate interplay between meningitis and mitochondria, identifying potential therapeutic interventions targeting mitochondria for the first time. With clinical trials on the horizon, our review lays the foundation for a transformative era in meningitis therapeutics, where unraveling the intricate interplay between meningitis and mitochondria offers promise for mitigating neuroinflammation and improving patient outcomes.
Collapse
Affiliation(s)
- Li-Li Wu
- Department of Encephalopathy, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China.
| | - Wei-Dong Shi
- Department of Orthopedics, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China.
| | - Wei-Feng Peng
- Department of Encephalopathy, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466000, China.
| | - Guo-Yin Li
- Department of Encephalopathy, Zhoukou Hospital of Traditional Chinese Medicine, Zhoukou 466099, China; College of Life Science and Agronomy, Zhoukou Normal University, Zhoukou 466000, China; Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi'an 710062, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
30
|
Gu H, Li Q, Liu Z, Li Y, Liu K, Kong X, Zhang Y, Meng Q, Song K, Xie Q, Gao Y, Cheng L. SPP1-ITGα5/β1 Accelerates Calcification of Nucleus Pulposus Cells by Inhibiting Mitophagy via Ubiquitin-Dependent PINK1/PARKIN Pathway Blockade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411162. [PMID: 39721032 PMCID: PMC11831503 DOI: 10.1002/advs.202411162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Low back pain (LBP) caused by nucleus pulposus degeneration and calcification leads to great economic and social burden worldwide. Unexpectedly, no previous studies have demonstrated the association and the underlying mechanism between nucleus pulposus tissue degeneration and calcification formation. Secreted Phosphoprotein 1 (SPP1) exerts crucial functions in bone matrix mineralization and calcium deposition. Here, a novel function of SPP1 is reported, namely that it can aggravate nucleus pulposus cells (NPs) degeneration by negatively regulating extracellular matrix homeostasis. The degenerated NPs have a higher mineralization potential, which is achieved by SPP1. Mechanistically, SPP1 can accelerate the degeneration of nucleus pulposus cells by activating integrin α5β1 (ITGα5/β1), aggravating mitochondrial damage and inhibiting mitophagy. SPP1-ITGα5/β1 axis inhibits mitophagy by PINK1/PARKIN pathway blockade. In conclusion, SPP1 activates ITGα5/β1 to inhibit mitophagy, accelerates NPs degeneration, and induces calcification, thereby leading to intervertebral disc degeneration (IVDD) and calcification, identifying the potentially unknown mechanism and relationship between IVDD and calcification. Important insights are provided into the role of SPP1 in nucleus pulposus calcification in IVDD by inducing nucleus pulposus cell senescence through inhibition of mitophagy and may help develop potential new strategies for IVDD treatment.
Collapse
Affiliation(s)
- Hanwen Gu
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qi Li
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Zhenchuan Liu
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Yanlin Li
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Kaiwen Liu
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Xiangzhen Kong
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Yuanqiang Zhang
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qunbo Meng
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Kangle Song
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Qing Xie
- Department of PharmacyQilu HospitalCheeloo College of MedicineShandong UniversityJinan250012China
| | - Yuan Gao
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| | - Lei Cheng
- Department of OrthopedicQilu Hospital of Shandong UniversityJinanShandong250012China
| |
Collapse
|
31
|
Lavrichenko DS, Chelebieva ES, Kladchenko ES. The mitochondrial membrane potential and the sources of reactive oxygen species in the hemocytes of the ark clam Anadara kagoshimensis under hypoosmotic stress. Comp Biochem Physiol B Biochem Mol Biol 2025; 276:111057. [PMID: 39662678 DOI: 10.1016/j.cbpb.2024.111057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
To compensate for changes in cell volume caused by changes in salt concentration, mollusks use regulatory mechanisms such as the regulation of volume decrease (RVD). This may increase the rate of aerobic metabolism and lead to an increase in reactive oxygen species (ROS). This study examined the production of ROS in the mitochondria of Anadara kagoshiensis hemocytes, the effect of mitochondrial inhibitors on osmotic stability in hemocytes, and the dynamics of changes in ROS levels and mitochondrial membrane potential when RVD is activated under hypo-osmotic conditions. Hemocytes maintained at a control osmolarity of 460 mOsm l-1 showed significant decreases in ROS production following incubation with complex III inhibitors (S3QEL). Hypoosmotic shock stimulated RVD in all experimental groups. The cell volume increased by about 70 % immediately after osmolarity was reduced, and then decreased by about 40 % over the next 30 min. A reduction in osmolarity from about 460 to 200 mOsm l-1 significantly decreased ROS and mitochondrial potentials in A. kashimensis hemocyctes. Inhibitors of mitochondrial complexes did not affect changes in ROS or mitochondria potentials in A kashimiensis hemocytes under hypoosmotic conditions or in hemocyte volume regulation mechanisms.
Collapse
Affiliation(s)
- Daria S Lavrichenko
- Laboratory of Ecological Immunology of Aquatic Organisms, A.O. Kovalevsky Institute of Biology of the Southern Seas of RAS, Leninsky Ave, 14, Moscow 119991, Russia.
| | - Elina S Chelebieva
- Laboratory of Ecological Immunology of Aquatic Organisms, A.O. Kovalevsky Institute of Biology of the Southern Seas of RAS, Leninsky Ave, 14, Moscow 119991, Russia
| | - Ekaterina S Kladchenko
- Laboratory of Ecological Immunology of Aquatic Organisms, A.O. Kovalevsky Institute of Biology of the Southern Seas of RAS, Leninsky Ave, 14, Moscow 119991, Russia
| |
Collapse
|
32
|
Yuan AT, Stillman MJ. Oxidative pathways of apo, partially, and fully Zn(II)- and Cd(II)-metalated human metallothionein-3 are dominated by disulfide bond formation. FEBS J 2025; 292:619-634. [PMID: 39617990 PMCID: PMC11796322 DOI: 10.1111/febs.17333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 09/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025]
Abstract
Oxidative stress is a key component of many diseases, including neurodegenerative diseases such as Alzheimer's disease. Reactive oxygen species (ROS) such as hydrogen peroxide and nitric oxide lead to disease progression by binding to proteins and causing their dysregulation. Metallothionein-3 (MT3), a cysteine-rich brain-located metalloprotein, has been proposed to be a key player in controlling oxidative stress in the central nervous system. We report data from a combination of electrospray ionization mass spectrometry (ESI-MS), ultraviolet (UV)-visible absorption spectroscopy, and circular dichroism spectroscopy that identify the oxidation pathway of MT3 fully bound to endogenous Zn(II) or exogenous Cd(II) together with the partially metalated species. We characterize the intermediate species formed during the oxidation of MT3, which is dominated by disulfide bond formation. We report the rates of oxidation. For both fully and partially metalated MT3, MT3 is oxidized at 5 to 10 times the rate of MT1, a similar but kidney-expressed isoform of MT. As oxidation progresses, MT3 follows a domain-specific demetallation pathway when it is fully metalated, and a domain-independent pathway when partially metalated. This suggests the presence of a significant susceptibility toward oxidation when MT3 is partially metalated, and, therefore, a possible protective role of Zn(II) when fully metalated. With the evidence for the rapid oxidation rate, our data support the proposals of MT3 as a key antioxidant in physiology.
Collapse
Affiliation(s)
- Amelia T. Yuan
- Department of ChemistryUniversity of Western OntarioLondonCanada
| | | |
Collapse
|
33
|
Moselhy OA, Abdel-Aziz N, El-Bahkery A, Moselhy SS, Ibrahim EA. Curcumin nanoparticles alleviate brain mitochondrial dysfunction and cellular senescence in γ-irradiated rats. Sci Rep 2025; 15:3857. [PMID: 39890961 PMCID: PMC11785741 DOI: 10.1038/s41598-025-87635-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Despite the diverse applications of γ radiation in radiotherapy, industrial processes, and sterilization, it causes hazardous effects on living organisms, such as cellular senescence, persistent cell cycle arrest, and mitochondrial dysfunction. This study evaluated the efficacy of curcumin nanoparticles (CNPs) in mitigating mitochondrial dysfunction and cellular senescence induced by γ radiation in rat brain tissues. Four groups of male Wistar albino rats (n = 8 per group) were included: (Gr1) the control group; (Gr2) the CNPs group (healthy rats receiving oral administration of curcumin nanoparticles at a dose of 10 mg/kg/day, three times per week for eight weeks); (Gr3) the irradiated group (rats exposed to a single dose of 10 Gy head γ irradiation); and (Gr4) the irradiated + CNPs group (irradiated rats treated with CNPs). The data obtained demonstrated that oral administration of CNPs for eight weeks attenuated oxidative stress in γ-irradiated rats by lowering the brain's lipid peroxidation level [malondialdehyde (MDA)] and enhancing antioxidant markers [superoxide dismutase (SOD), reduced glutathione (GSH), and total antioxidant capacity (TAC)] (P < 0.05). In addition, CNPs significantly increased mitochondrial function by improving complex I, complex II, and ATP production levels compared to the irradiated group. In irradiated rats, CNPs also showed anti-neuroinflammatory effects by reducing brain interleukin 6 (IL-6), tumor necrosis factor-alpha (TNF-α), and nuclear factor-kappa B (NF-ĸB) levels (P < 0.05). Moreover, CNPs administered to irradiated rats significantly reduced brain β-galactosidase activity and the expression levels of p53, p21, and p16 genes (P < 0.05) while concurrently inducing a significant increase in AMPK mRNA expression compared to the irradiated group. In conclusion, CNPs ameliorated the neurotoxicity of γ radiation and hold promise as a novel agent to delay cellular senescence via their combined antioxidant, anti-inflammatory, and mitochondrial-enhancing properties.
Collapse
Affiliation(s)
- Omnia A Moselhy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nahed Abdel-Aziz
- Radiation Biology Research Department, National Center for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Azza El-Bahkery
- Radiation Biology Research Department, National Center for Radiation Research & Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Said S Moselhy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Ehab A Ibrahim
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
34
|
Zheng C, Hu X, Hua R, Ren X, Shi S, Hong X, Wang Y, Qiu L, Wu D, Cao T, Huang S, Zhao S, Pan Y. A Cerium Oxide Loaded Hyaluronic Acid Nanosystem Remits Glucose Oxidative Stress-Induced Odontoblasts Mitochondrial Apoptosis through Regulation of PGAM5 Pathway. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4426-4439. [PMID: 39772475 DOI: 10.1021/acsami.4c13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Diabetes mellitus (DM) induced mitochondrial oxidative stress (OS) can lead to severe injury of dental pulp. The cerium oxide nanoparticles (CNP) have been proven to have excellent antioxidative activity. However, whether CNP can relieve dental pulp damage caused by DM and the underlying mechanisms remain unclear. In this study, we modified ceria with hyaluronic acid to prepare nanoceria with good biocompatibility, water solubility, and stability, namely, HACNP (hyaluronic acid cerium oxide nanoparticles). We demonstrated the protective effect of HACNP on diabetic OS-induced mitochondrial apoptosis in dental pulp-like cells. As far as the mechanism of action was concerned, glucose oxidase (GO) treatment promoted the activation of phosphoglycerate mutase family 5 (PGAM5) leading to mitochondrial abnormalities and apoptosis in an odontoblast-like cell line (mDPC6T). Knockdown or overexpression of PGAM5 further validate these results. Meanwhile, HACNP remitted GO-related toxicity via down-regulating PGAM5 expression, whereas overexpression of PGAM5 abolished the beneficial effect of HACNP. Furthermore, in the constructed animal research model of diabetic pulp injury, we also confirmed that HACNP alleviated apoptosis and mitochondrial injury of dental pulp and decreased the expression level of PGAM5 in diabetic pulp tissue. In conclusion, these results revealed that HACNP played a protective role on diabetes-associated dental pulp injury through targeting the PGAM5-mediated mitochondrial pathway, providing an idea and method for the prevention or treatment of diabetes-induced dental pulp damage.
Collapse
Affiliation(s)
- Chuchu Zheng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiangyu Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Ruize Hua
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Xuekun Ren
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Shuai Shi
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xinhua Hong
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Yilin Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Lili Qiu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Danni Wu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Tong Cao
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang LaboratoryZhejiang Lab for Regenerative Medicine, Vision, and Brain Health, Wenzhou 325027, China
| | - Shengbin Huang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Shufan Zhao
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Yihuai Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
35
|
Wen H, Deng H, Li B, Chen J, Zhu J, Zhang X, Yoshida S, Zhou Y. Mitochondrial diseases: from molecular mechanisms to therapeutic advances. Signal Transduct Target Ther 2025; 10:9. [PMID: 39788934 PMCID: PMC11724432 DOI: 10.1038/s41392-024-02044-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/28/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
Mitochondria are essential for cellular function and viability, serving as central hubs of metabolism and signaling. They possess various metabolic and quality control mechanisms crucial for maintaining normal cellular activities. Mitochondrial genetic disorders can arise from a wide range of mutations in either mitochondrial or nuclear DNA, which encode mitochondrial proteins or other contents. These genetic defects can lead to a breakdown of mitochondrial function and metabolism, such as the collapse of oxidative phosphorylation, one of the mitochondria's most critical functions. Mitochondrial diseases, a common group of genetic disorders, are characterized by significant phenotypic and genetic heterogeneity. Clinical symptoms can manifest in various systems and organs throughout the body, with differing degrees and forms of severity. The complexity of the relationship between mitochondria and mitochondrial diseases results in an inadequate understanding of the genotype-phenotype correlation of these diseases, historically making diagnosis and treatment challenging and often leading to unsatisfactory clinical outcomes. However, recent advancements in research and technology have significantly improved our understanding and management of these conditions. Clinical translations of mitochondria-related therapies are actively progressing. This review focuses on the physiological mechanisms of mitochondria, the pathogenesis of mitochondrial diseases, and potential diagnostic and therapeutic applications. Additionally, this review discusses future perspectives on mitochondrial genetic diseases.
Collapse
Affiliation(s)
- Haipeng Wen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Hui Deng
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Xian Zhang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Kurume, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
36
|
Fila M, Przyslo L, Derwich M, Pawlowska E, Blasiak J. Sexual Dimorphism in Migraine. Focus on Mitochondria. Curr Pain Headache Rep 2025; 29:11. [PMID: 39760955 DOI: 10.1007/s11916-024-01317-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE OF REVIEW Migraine prevalence in females is up to 3 times higher than in males and females show higher frequency, longer duration, and increased severity of headache attacks, but the reason for that difference is not known. This narrative review presents the main aspects of sex dimorphism in migraine prevalence and discusses the role of sex-related differences in mitochondrial homeostasis in that dimorphism. The gender dimension is also shortly addressed. RECENT FINDINGS The imbalance between energy production and demand in the brain susceptible to migraine is an important element of migraine pathogenesis. Mitochondria are the main energy source in the brain and mitochondrial impairment is reported in both migraine patients and animal models of human migraine. However, it is not known whether the observed changes are consequences of primary disturbance of mitochondrial homeostasis or are secondary to the migraine-affected hyperexcitable brain. Sex hormones regulate mitochondrial homeostasis, and several reports suggest that the female hormones may act protectively against mitochondrial impairment, contributing to more effective energy production in females, which may be utilized in the mechanisms responsible for migraine progression. Migraine is characterized by several comorbidities that are characterized by sex dimorphism in their prevalence and impairments in mitochondrial functions. Mitochondria may play a major role in sexual dimorphism in migraine through the involvement in energy production, the dependence on sex hormones, and the involvement in sex-dependent comorbidities. Studies on the role of mitochondria in sex dimorphism in migraine may contribute to precise personal therapeutic strategies.
Collapse
Affiliation(s)
- Michal Fila
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| | - Lukasz Przyslo
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| | - Marcin Derwich
- Department of Developmental Dentistry, Medical University of Lodz, 90-647, Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Developmental Dentistry, Medical University of Lodz, 90-647, Lodz, Poland
| | - Janusz Blasiak
- Faculty of Medicine, Collegium Medicum, Mazovian Academy in Plock, 09-420, Plock, Poland.
| |
Collapse
|
37
|
Xu L, Li C, Wan T, Sun X, Lin X, Yan D, Li J, Wei P. Targeting uric acid: a promising intervention against oxidative stress and neuroinflammation in neurodegenerative diseases. Cell Commun Signal 2025; 23:4. [PMID: 39754256 PMCID: PMC11699683 DOI: 10.1186/s12964-024-01965-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/27/2024] [Indexed: 01/06/2025] Open
Abstract
Oxidative stress and neuroinflammation are recognized as key factors in the development of neurodegenerative diseases, yet effective interventions and biomarkers to address oxidative stress and neuroinflammation in these conditions are limited. Uric acid (UA), traditionally associated with gout, is now gaining prominence as a potential target in neurodegenerative diseases. Soluble UA stands out as one of the most vital antioxidant compounds produced by the human body, accounting for up to 55% of the extracellular capacity to neutralize free radicals. While there is increasing evidence supporting the neuroprotective properties of UA in Parkinson's disease and Alzheimer's disease, gaps in knowledge still exist regarding the underlying mechanisms and how to effectively translate these benefits into clinical practice. Moreover, the current UA elevation therapy exhibits unstable antioxidant properties, individual variability, and even adverse effects, limiting its potential clinical applications. This review consolidates recent advancements in understanding how UA exerts neuroprotective effects on neurodegenerative diseases and emphasizes the dual roles of UA in managing oxidative stress and neuroinflammation. Additionally, the review elucidates the mechanisms through which UA confers neuroprotection. Based on this, the review underscores the significance of UA as a potential biomarker and aims to provide a comprehensive understanding of its potential as a therapeutic target, while also addressing possible challenges to clinical implementation.
Collapse
Affiliation(s)
- Lin Xu
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Chengwei Li
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Tiantian Wan
- Department of Anesthesiology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xinyi Sun
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Xiaojie Lin
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Dong Yan
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Jianjun Li
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China
| | - Penghui Wei
- Department of Anesthesiology, Cheeloo College of Medicine, Qilu Hospital (Qingdao), Shandong University, 758 Hefei Road, Qingdao, China.
- Laboratory of Anesthesia and Brain Function, Qilu hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, China.
| |
Collapse
|
38
|
Lilley T, Camera DM, Kwa FAA. Repairing muscle with broccoli-derived sulforaphane: A preclinical evaluation for the treatment of mitochondrial myopathies. Drug Discov Today 2025; 30:104283. [PMID: 39736463 DOI: 10.1016/j.drudis.2024.104283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
Skeletal muscle health relies on the production of adenosine triphosphate (ATP) in the mitochondria. ATP production is accompanied by oxidative phosphorylation, which generates reactive oxygen species (ROS). When there is an imbalance in ROS levels, oxidative stress and subsequent mitochondrial dysfunction, mitochondrial myopathies including sarcopenia, chronic progressive external ophthalmoplegia, and proximal myopathy can result. Such incurable myopathies are characterised by aberrant metabolism, limited ATP production, and muscle atrophy. Broccoli-derived sulforaphane has emerged as a novel treatment for mitochondrial myopathies because of its antioxidant and anti-inflammatory properties. This review discusses preclinical studies that reveal sulforaphane's potential therapeutic benefits and limitations in treating mitochondrial myopathies.
Collapse
Affiliation(s)
- Thomas Lilley
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Donny M Camera
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia
| | - Faith A A Kwa
- Department of Biomedical, Health and Exercise Sciences, Swinburne University of Technology, Melbourne, Australia.
| |
Collapse
|
39
|
Jayaweera SW, Sahin M, Lundkvist F, Leven A, Tereenstra L, Bäckman J, Bachhar A, Bano F, Anan I, Olofsson A. Misfolding of transthyretin in vivo is controlled by the redox environment and macromolecular crowding. J Biol Chem 2025; 301:108031. [PMID: 39615680 PMCID: PMC11732491 DOI: 10.1016/j.jbc.2024.108031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Transthyretin (TTR) amyloidosis is a progressive disorder characterized by peripheral neuropathy, autonomic dysfunction, and cardiomyopathy. The precise mechanism by which TTR misfolds and forms fibrils in vivo remains incompletely understood, posing challenges to the development of effective therapeutics. In this study, we reveal that the recently identified nonnative pathological species of TTR (NNTTR), which is enriched in the plasma of ttr-val30met gene carriers, exhibits strong amyloidogenic properties, making it a promising therapeutic target. Notably, we demonstrate that NNTTR formation is dependent on an intermolecular disulfide bond and can be promoted by oxidative conditions while being effectively suppressed by reducing agents. The formation of this disulfide bond is incompatible with the native TTR fold, thereby necessitating structural flexibility. We further show that this required flexibility can be constrained using tetramer-stabilizing drugs, thereby suppressing NNTTR formation. Interestingly, the flexibility is also hindered by macromolecular crowding, and NNTTR formation is strongly suppressed by the high protein concentration in plasma. This suppression is released upon dilution, which thus promotes NNTTR formation in areas with lower protein content, highlighting a potential link to the interstitial space, brain, and vitreous body of the eye, where TTR-amyloid is frequently observed. In summary, we demonstrate that NNTTR displays strong amyloidogenic features, underscoring its potential as a therapeutic target. We identify the redox environment and macromolecular crowding as key modulatory factors. Our findings propose a mechanistic explanation for TTR misfolding and suggest a novel therapeutic approach.
Collapse
Affiliation(s)
| | - Melisnur Sahin
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Fabian Lundkvist
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Alice Leven
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Laura Tereenstra
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Joel Bäckman
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Anushree Bachhar
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Fouzia Bano
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Intissar Anan
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Anders Olofsson
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden.
| |
Collapse
|
40
|
Jolivet N, Bertolin G. Revealing mitochondrial architecture and functions with single molecule localization microscopy. Biol Cell 2025; 117:e2400082. [PMID: 39877953 PMCID: PMC11775716 DOI: 10.1111/boc.202400082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/31/2025]
Abstract
Understanding the spatiotemporal organization of components within living systems requires the highest resolution possible. Microscopy approaches that allow for a resolution below 250 nm include electron and super-resolution microscopy (SRM). The latter combines advanced imaging techniques and the optimization of image processing methods. Over the last two decades, various SRM-related approaches have been introduced, especially those relying on single molecule localization microscopy (SMLM). To develop and apply SMLM approaches, mitochondria are an ideal cellular compartment due to their size, which is below the standard diffraction limit. Furthermore, mitochondria are a dynamic yet narrow compartment, and a resolution below 250 nm is required to study their composition and multifaceted functions. To this end, several SMLM technologies have been used to reveal mitochondrial composition. However, there is still room for improvement in existing techniques to study protein-protein interactions and protein dynamics within this compartment. This review aims to offer an updated overview of the existing SMLM techniques and probes associated with mitochondria to enhance their resolution at the nanoscale. Last, it paves the way for future SMLM improvements to better resolve mitochondrial dynamics and functions.
Collapse
Affiliation(s)
- Nicolas Jolivet
- CNRSUniv Rennes, IGDR [(Institut de Génétique et Développement de Rennes)]‐UMR 6290RennesFrance
| | - Giulia Bertolin
- CNRSUniv Rennes, IGDR [(Institut de Génétique et Développement de Rennes)]‐UMR 6290RennesFrance
| |
Collapse
|
41
|
Zhang N, Nao J, Dong X. Efficacy and Safety of Natural Apigenin Treatment for Alzheimer's Disease: Focus on In vivo Research Advancements. Curr Neuropharmacol 2025; 23:728-754. [PMID: 39665306 DOI: 10.2174/1570159x23666241211095018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most common dementia in clinics. Despite decades of progress in the study of the pathogenesis of AD, clinical treatment strategies for AD remain lacking. Apigenin, a natural flavonoid compound, is present in a variety of food and Chinese herbs and has been proposed to have a wide range of therapeutic effects on dementia. OBJECTIVE To clarify the relevant pharmacological mechanism and therapeutic effect of apigenin on animal models of AD. METHODS Computer-based searches of the PubMed, Cochrane Library, Embase, and Web of Science databases were used to identify preclinical literature on the use of apigenin for treating AD. All databases were searched from their respective inception dates until June 2023. The meta-analysis was performed with Review manager 5.4.1 and STATA 17.0. RESULTS Thirteen studies were eventually enrolled, which included 736 animals in total. Meta-analysis showed that apigenin had a positive effect on AD. Compared to controls, apigenin treatment reduced escape latency, increased the percentage of time spent in the target quadrant and the number of plateaus traversed; apigenin was effective in reducing nuclear factor kappa-B (NF-κB) p65 levels; apigenin effectively increased antioxidant molecules SOD and GSH-px and decreased oxidative index MDA; for ERK/CREB/BDNF pathway, apigenin effectively increased BDNF and pCREB molecules; additionally, apigenin effectively decreased caspase3 levels and the number of apoptotic cells in the hippocampus. CONCLUSION The results show some efficacy of apigenin in the treatment of AD models. However, further clinical studies are needed to confirm the clinical efficacy of apigenin.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neurology, China Medical University Seventh Clinical College, Fushun, Liaoning P.R. China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
42
|
Wang TY, Yang Q, Cheng XY, Ding JC, Hu PF. Beyond weight loss: the potential of glucagon-like peptide-1 receptor agonists for treating heart failure with preserved ejection fraction. Heart Fail Rev 2025; 30:17-38. [PMID: 39269643 DOI: 10.1007/s10741-024-10438-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2024] [Indexed: 09/15/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with various phenotypes, and obesity is one of the most common and clinically relevant phenotypes of HFpEF. Obesity contributes to HFpEF through multiple mechanisms, including sodium retention, neurohormonal dysregulation, altered energy substrate metabolism, expansion of visceral adipose tissue, and low-grade systemic inflammation. Glucagon-like peptide-1 (GLP-1) is a hormone in the incretin family. It is produced by specialized cells called neuroendocrine L cells located in the distal ileum and colon. GLP-1 reduces blood glucose levels by promoting glucose-dependent insulin secretion from pancreatic β cells, suppressing glucagon release from pancreatic α cells, and blocking hepatic gluconeogenesis. Recent evidence suggests that GLP-1 receptor agonists (GLP-1 RAs) can significantly improve physical activity limitations and exercise capacity in obese patients with HFpEF. The possible cardioprotective mechanisms of GLP-1 RAs include reducing epicardial fat tissue thickness, preventing activation of the renin-angiotensin-aldosterone system, improving myocardial energy metabolism, reducing systemic inflammation and cardiac oxidative stress, and delaying the progression of atherosclerosis. This review examines the impact of obesity on the underlying mechanisms of HFpEF, summarizes the trial data on cardiovascular outcomes of GLP-1 RAs in patients with type 2 diabetes mellitus, and highlights the potential cardioprotective mechanisms of GLP-1 RAs to give a pathophysiological and clinical rationale for using GLP-1 RAs in obese HFpEF patients.
Collapse
Affiliation(s)
- Tian-Yu Wang
- Department of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Yang
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin-Yi Cheng
- Department of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun-Can Ding
- Department of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peng-Fei Hu
- Department of Cardiology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
43
|
Zhou Z, Li Y, Ding J, Sun S, Cheng W, Yu J, Cai Z, Ni Z, Yu C. Chronic unpredictable stress induces anxiety-like behavior and oxidative stress, leading to diminished ovarian reserve. Sci Rep 2024; 14:30681. [PMID: 39730417 DOI: 10.1038/s41598-024-76717-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/16/2024] [Indexed: 12/29/2024] Open
Abstract
Chronic stress can adversely affect the female reproductive endocrine system, potentially leading to disorders and impairments in ovarian function. However, current research lacks comprehensive understanding regarding the biochemical characteristics and underlying mechanisms of ovarian damage induced by chronic stress. We established a stable chronic unpredictable stress (CUS)-induced diminished ovarian reserve (DOR) animal model. Our findings demonstrated that prolonged CUS treatment over eight weeks resulted in increased atresia follicles in female mice. This atresia was accompanied by decreased AMH and increased FSH levels. Furthermore, we observed elevated levels of corticosterone both in the peripheral blood and within the ovary. Additionally, we detected abnormalities in ATP metabolism within the ovarian tissue. CUS exposure led to oxidative stress in the ovaries, fostering a microenvironment characterized by oxidative damage to mouse ovarian granulosa cells (mGCs) and heightened levels of reactive oxygen species. Furthermore, CUS prompted mGCs to undergo apoptosis via the mitochondrial pathway. These findings indicate a direct association between the fundamental physiological alterations leading to DOR and the oxidative phosphorylation processes within mGCs. The diminished ATP production by mGCs, triggered by CUS, emerges as a pivotal indicator of CUS-induced DOR. Our study establishes an animal model to investigate the impact of chronic stress on ovarian reserve function and sheds light on potential mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Zhihao Zhou
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China
- Traditional Chinese Medicine Department, No. 929 Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yangshuo Li
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China
| | - Jie Ding
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China
| | - Shuai Sun
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China
| | - Wen Cheng
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China
| | - Jin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China
| | - Zailong Cai
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai, 200433, China
| | - Zhexin Ni
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China.
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, 100850, China.
| | - Chaoqin Yu
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Military Medical University (Changhai Hospital), 168 Changhai Road, Yangpu District, Shanghai, China.
| |
Collapse
|
44
|
Li P, Zhou M, Wang J, Tian J, Zhang L, Wei Y, Yang F, Xu Y, Wang G. Important Role of Mitochondrial Dysfunction in Immune Triggering and Inflammatory Response in Rheumatoid Arthritis. J Inflamm Res 2024; 17:11631-11657. [PMID: 39741752 PMCID: PMC11687318 DOI: 10.2147/jir.s499473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 12/15/2024] [Indexed: 01/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory autoimmune disease, primarily characterized by chronic symmetric synovial inflammation and erosive bone destruction.Mitochondria, the primary site of cellular energy production, play a crucial role in energy metabolism and possess homeostatic regulation capabilities. Mitochondrial function influences the differentiation, activation, and survival of both immune and non-immune cells involved in RA pathogenesis. If the organism experiences hypoxia, genetic predisposition, and oxidative stress, it leads to mitochondrial dysfunction, which further affects immune cell energy metabolism, synovial cell proliferation, apoptosis, and inflammatory signaling, causing the onset and progression of RA; and, mitochondrial regulation is becoming increasingly important in the treatment of RA.In this review, we examine the structure and function of mitochondria, analyze the potential causes of mitochondrial dysfunction in RA, and focus on the mechanisms by which mitochondrial dysfunction triggers chronic inflammation and immune disorders in RA. We also explore the effects of mitochondrial dysfunction on RA immune cells and osteoblasts, emphasizing its key role in the immune response and inflammatory processes in RA. Furthermore, we discuss potential biological processes that regulate mitochondrial homeostasis, which are of great importance for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Pingshun Li
- College of Integrative Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Mengru Zhou
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Jia Wang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Jiexiang Tian
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Lihuan Zhang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Yong Wei
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Fang Yang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Yali Xu
- College of Integrative Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| | - Gang Wang
- Department of Rheumatology and Bone Disease, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, People’s Republic of China
| |
Collapse
|
45
|
Borisenkova AA, Eropkin MY, Konovalova NI, Titova AV, Markova MA, Lyutova ZB, Mazur AS, Sedov VP, Orlova VA, Lykholay AN, Orlova DN, Arutyunyan AV. Fullerenol C 60(OH) 36: Antioxidant, Cytoprotective, Anti-Influenza Virus Activity, and Self-Assembly in Aqueous Solutions and Cell Culture Media. Antioxidants (Basel) 2024; 13:1525. [PMID: 39765853 PMCID: PMC11727559 DOI: 10.3390/antiox13121525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/15/2025] Open
Abstract
Viral infections and many other dangerous diseases are accompanied by the development of oxidative stress, which is a consequence of an increase in the level of the reactive oxygen species (ROS). In this regard, the search for effective antioxidants remains highly relevant. We tested fullerenol C60(OH)36 in the context of the connection between its self-assembly in aqueous solutions and cell culture media, antiradical activity, UV cytoprotective action, and antiviral activity against international reference strains of influenza virus A(H1N1)pdm09, A(H3N2), and B subtypes in vitro on the MDCK cell line. Various characterization techniques, including Fourier-transform infrared spectroscopy (FTIR), Raman spectroscopy, NMR and ESR spectrometry, MALDI-TOF mass spectrometry, thermal analysis (TGA and DSC), dynamic light-scattering (DLS), and ζ-potential measurements, were used to confirm the production of fullerenol and study its self-assembly in aqueous solutions and cell culture media. Fullerenol C60(OH)36 demonstrated the ability to scavenge •DPPH, •OH, O2•- radicals and 1O2 and was non-toxic in the range of the studied concentrations (up to 200 μg/mL) when incubated with MDCK cells for 24 h. In addition, fullerenol exhibited a cytoprotective effect under UV irradiation (EC50 = 29.7 ± 1.0 μM) and showed moderate activity against human influenza viruses of subtypes A(H1N1)pdm09 (SI = 9.9 ± 4.6) and A(H3N2) (SI = 12.5 ± 1.3) when determined by the hemagglutination assay (HA-test) and the MTT assay. At the same time, C60(OH)36 was ineffective in vitro against the actual strain of influenza B virus (Victoria lineage). The high bioavailability of fullerenol in combination with its cytoprotective effect, as well as its antiradical and antiviral activity combined with a relatively low toxicity, allows to consider it a promising compound for biomedical applications.
Collapse
Affiliation(s)
- Alina A. Borisenkova
- Radiation Technology Department, St. Petersburg State Institute of Technology, Technical University, 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Mikhail Y. Eropkin
- Smorodintsev Research Institute of Influenza, 197022 St. Petersburg, Russia
| | | | - Anna V. Titova
- Radiation Technology Department, St. Petersburg State Institute of Technology, Technical University, 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Maria A. Markova
- Radiation Technology Department, St. Petersburg State Institute of Technology, Technical University, 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Zhanna B. Lyutova
- Radiation Technology Department, St. Petersburg State Institute of Technology, Technical University, 190013 St. Petersburg, Russia
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Anton S. Mazur
- Research Resource Center “Magnetic Resonance Research Methods”, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Victor P. Sedov
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | | | - Anna N. Lykholay
- Research Resource Center “Molecular and Cell Technologies”, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Diana N. Orlova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| | - Alexandr V. Arutyunyan
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre “Kurchatov Institute”, 188300 Gatchina, Russia
| |
Collapse
|
46
|
Qin L, Huang T, Zhang D, Wei L, Li G, Zhu Q, Tong Q, Ding G, Liu J. The mitochondrial function of peripheral blood cells in cognitive frailty patients. Front Aging Neurosci 2024; 16:1503246. [PMID: 39723155 PMCID: PMC11669044 DOI: 10.3389/fnagi.2024.1503246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Background Cognitive frailty (CF), characterized by the coexistence of physical frailty and cognitive impairment, is linked to increased morbidity and mortality in older adults. While CF has been linked to multiple physiological and lifestyle factors, the underlying biological mechanisms remain poorly understood. This study investigated the risk factors for CF and explored the relationship between mitochondrial function and CF in hospitalized patients. Methods A total of 279 hospitalized individuals were recruited from December 2020 to August 2022, conducted comprehensive clinical assessments, and collected peripheral blood samples. CF was evaluated using the Physical Frailty Phenotype and Montreal Cognitive Assessment scales. Nutritional status was assessed with the Mini Nutritional Assessment, and depression was measured using the Geriatric Depression Scale. DNA was obtained from the peripheral blood and interrogated for mitochondrial DNA copy number (mtDNAcn). Peripheral blood mononuclear cells isolated from peripheral blood were examined for respiratory function and reactive oxygen species (ROS) levels. Additionally, plasma samples were analyzed for inflammatory markers and Carnitine Palmitoyltransferase II (CPT2). Results Among the participants, 90 were classified as CF and 46 as non-CF. Logistic regression analysis revealed that increased age (OR 1.156, 95% CI 1.064-1.255), lower educational attainment (OR 0.115, 95% CI 0.024-0.550), malnutrition (OR 0.713, 95% CI 0.522-0.973), and higher depression scores (OR 1.345, 95% CI 1.065-1.699) were significantly associated with CF. The independent t tests and Mann-Whitney U tests showed the CF group exhibited impaired mitochondrial function, characterized by reduced mtDNAcn and respiratory activity, coupled with elevated ROS, interleukin-6, and CPT2 levels compared with the non-CF group. After adjusted for age, sex, and BMI, compared with non-CF group, the OR values for the CF group of mtDNAcn and ROS were 0.234 (95% CI = 0.065-0.849) (p = 0.027) and 1.203 (95% CI = 1.075-1.347) (p = 0.001), respectively. The Sensitive analysis showed that the area under curve values for mtDNAcn and ROS were 0.653 and 0.925. Conclusion Age, lower educational attainment, malnutrition, and depression are significant risk factors for CF. Moreover, mitochondrial dysfunction, characterized by decreased mtDNAcn, impaired respiratory function and increased ROS levels appears to be a critical phenotype of CF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Guoxian Ding
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Juan Liu
- Division of Geriatric Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
47
|
Shao N, Lu Q, Ouyang Z, Yang P, Wei T, Wang J, Cai B. Ganoderic acid a alleviates Aβ 25-35-induced HT22 cell apoptosis through the ERK/MAPK pathway: a system pharmacology and in vitro experimental validation. Metab Brain Dis 2024; 40:51. [PMID: 39625499 DOI: 10.1007/s11011-024-01429-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/31/2024] [Indexed: 02/27/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that occurs with aging. Ganoderma lucidum (Curtis.) P. Karst. (G. lucidum) is a traditional medicinal fungus believed to nourish the brain and anti-aging. Ganoderic acid A (GAA), a triterpenoid from G. lucidum, has demonstrated natural neuroprotective effects. This study aims to explore the therapeutic effect and molecular mechanism of GAA on AD. Systematic network pharmacology identified 95 targets, 8 biological functions, and multiple pathways. The results highlighted MAPK family members as core genes, with MAPK1 (ERK2) showing the highest binding affinity to GAA in molecular docking. In vitro experiments revealed that GAA dose-dependently increased the viability of Aβ25-35-injured HT22 cells and inhibited MAPK pathway-related protein expression. Similar to FR180204, 100 µM GAA significantly reversed ERK protein expression, oxidative stress markers, and mitochondrial damage in AD cell model. GAA also downregulated cleaved caspase-3 protein levels, apoptosis rates, Aβ and p-Tau expression by inhibiting the ERK signaling pathway. The therapeutic effect of GAA on AD was predicted and validated through network pharmacology and in vitro experiments. The ability of GAA to inhibit apoptosis via the ERK/MAPK signaling pathway positions it as a promising candidate for AD treatment.
Collapse
Affiliation(s)
- Nan Shao
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Qingyang Lu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhaorong Ouyang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Peizheng Yang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Tao Wei
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Jinghui Wang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China.
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China.
| |
Collapse
|
48
|
Gao S, Cheng Q, Hu Y, Fan X, Liang C, Niu C, Kang Q, Wei T. Melatonin antagonizes oxidative stress-induced apoptosis in retinal ganglion cells through activating the thioredoxin-1 pathway. Mol Cell Biochem 2024; 479:3393-3404. [PMID: 38353878 DOI: 10.1007/s11010-024-04924-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 01/05/2024] [Indexed: 03/28/2024]
Abstract
This study aimed to explore the role of melatonin in oxidative stress-induced injury on retinal ganglion cells and the underlying mechanisms. The immortalized RGC-5 cells were treated with H2O2 to induce oxidative injury. Cell viability was measured by Cell Counting Kit-8, and apoptosis was determined by flow cytometry and western blot assays. Reactive oxygen species (ROS), lactate dehydrogenase (LDH), and malondialdehyde (MDA) levels were examined to evaluate oxidative stress levels. In addition, Thioredoxin-1 (Trx1) was silenced in RGC-5 cells using small interfering RNA followed by signaling pathway examination to explore the underlying mechanisms of melatonin in alleviating oxidative injury. Melatonin pre-treatment significantly alleviated H2O2-induced apoptosis in RGC-5 cells. Melatonin also markedly reversed the upregulation of cleaved-caspase 3, cleaved-caspase 9, and Bax expression and downregulation of Bcl-2 expression induced by H2O2. Further analyses presented that melatonin significantly attenuated the increase of ROS, LDH, and MDA levels in RGC-5 cells after H2O2 treatment. Melatonin also abolished the downregulated expression of Superoxide dismutase type 1, Trx1, and Thioredoxin reductase 1, and the reduced activity of thioredoxin reductase in RGC-5 cells after H2O2 treatment. Notably, Trx1 knockdown significantly mitigated the protective effect of melatonin in alleviating H2O2-induced apoptosis and oxidative stress, while administration of compound C, a common inhibitor of c-Jun N-terminal kinase (JNK) signaling, partially reversed the effect of Trx1 silencing, thereby ameliorating the apoptosis and oxidative injury induced by H2O2 in RGC-5 cells. Melatonin could significantly alleviate oxidative stress-induced injury of retinal ganglion cells via modulating Trx1-mediated JNK signaling pathway.
Collapse
Affiliation(s)
- Shan Gao
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Qiaochu Cheng
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Yaguang Hu
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Xiaojuan Fan
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Chen Liang
- Department of Neurosurgery, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Chen Niu
- Department of Medical Imaging, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Qianyan Kang
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China
| | - Ting Wei
- Department of Ophthalmology, the First Affiliated Hospital of Xi'an Jiaotong University, No.277 Yanta West Road, Yanta District, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
49
|
Bai X, Lu H, Cui Y, Yu S, Ma R, Yang S, He J. PRKAA2-mediated mitophagy regulates oxygen consumption in yak renal tubular epithelial cells under chronic hypoxia. Cell Signal 2024; 124:111450. [PMID: 39396565 DOI: 10.1016/j.cellsig.2024.111450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Hypoxic environments are significant factors in the induction of various kidney diseases and are closely associated with high oxygen consumption in the kidneys. Yaks live at high altitude for a long time, exhibit a unique ability to regulate kidney oxygen consumption, protecting them from hypoxia-induced damage. However, the mechanisms underlying the regulation of oxygen consumption in yak kidneys under hypoxic conditions remain unclear. To explore this hypoxia adaptation mechanism in yak kidneys, this study analyzed the oxygen consumption rate (OCR) of renal tubular epithelial cells (RTECs) under hypoxia. We found that the OCR and apoptosis rates of RTECs under chronic hypoxia (> 24 h) were lower than those under acute hypoxia (≤ 24 h). However, when oxygen consumption was promoted under chronic hypoxia, the apoptosis rate increased, indicating that reducing the cellular OCR is crucial for maintaining RTECs activity under hypoxia. High-throughput sequencing results showed that the mitophagy pathway is likely a key mechanism for inhibiting OCR of yak RTECs, with protein kinase AMP-activated catalytic subunit alpha 2 (PRKAA2) playing a significant role in this process. Further studies demonstrated that chronic hypoxia activates the mitophagy pathway, which inhibits oxidative phosphorylation (OXPHOS) while increasing glycolytic flux in yak RTECs. Conversely, when the mitophagy pathway was inhibited, there was an increase in the activity of OXPHOS enzymes and OCR. To further explore the role of PRKAA2 in the mitophagy pathway, we inhibited PRKAA2 expression under chronic hypoxia. Results showed that the downregulation of PRKAA2 decreased the expression of mitophagy-related proteins, such as p-FUNDC1/FUNDC1, LC3-II/LC3-I, BNIP3 and ULK1 while upregulating P62 expression. Additionally, there was an increase in the enzyme activities of Complex II, Complex IV, PDH, and SDH, which further promoted oxygen consumption in RTECs. These findings suggest that PRKAA2 mediated mitophagy under chronic hypoxia is crucial mechanism for reducing oxygen consumption in yak RTECs.
Collapse
Affiliation(s)
- Xuefeng Bai
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Hongqin Lu
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China.
| | - Sijiu Yu
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Rui Ma
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Shanshan Yang
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Junfeng He
- Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
50
|
Huang Y, Li W, Sun H, Guo X, Zhou Y, Liu J, Liu F, Fan Y. Mitochondrial transfer in the progression and treatment of cardiac disease. Life Sci 2024; 358:123119. [PMID: 39395616 DOI: 10.1016/j.lfs.2024.123119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/27/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
Mitochondria are the primary site for energy production and play a crucial role in supporting normal physiological functions of the human body. In cardiomyocytes (CMs), mitochondria can occupy up to 30 % of the cell volume, providing sufficient energy for CMs contraction and relaxation. However, some pathological conditions such as ischemia, hypoxia, infection, and the side effect of drugs, can cause mitochondrial dysfunction in CMs, leading to various myocardial injury-related diseases including myocardial infarction (MI), myocardial hypertrophy, and heart failure. Self-control of mitochondria quality and conversion of metabolism pathway in energy production can serve as the self-rescue measure to avoid autologous mitochondrial damage. Particularly, mitochondrial transfer from the neighboring or extraneous cells enables to mitigate mitochondrial dysfunction and restore their biological functions in CMs. Here, we described the homeostatic control strategies and related mechanisms of mitochondria in injured CMs, including autologous mitochondrial quality control, mitochondrial energy conversion, and especially the exogenetic mitochondrial donation. Additionally, this review emphasizes on the therapeutic effects and potential application of utilizing mitochondrial transfer in reducing myocardial injury. We hope that this review can provide theoretical clues for the developing of advanced therapeutics to treat cardiac diseases.
Collapse
Affiliation(s)
- Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Hongyu Sun
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Xin Guo
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| | - Yonghong Fan
- The General Hospital of Western Theater Command, Chengdu 610083, China.
| |
Collapse
|