1
|
Ni W, Chen SH, Dai L, Zhou Y, He C. A prospective cohort study on serum PINK1 as a biochemical marker in relation to poor neurological prognosis, stroke-associated pneumonia and early neurological deterioration after acute intracerebral hemorrhage. Clin Chim Acta 2025; 573:120282. [PMID: 40199435 DOI: 10.1016/j.cca.2025.120282] [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: 01/06/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND PTEN-induced putative kinase 1 (PINK1) may moderate neurodegeneration via sustaining mitochondrial function and integrity. Here, we attempted to determine relationship between serum PINK1 levels, disease severity, poor prognosis, Early Neurological Deterioration (END) and Stroke-Associated Pneumonia (SAP) following acute Intracerebral Hemorrhage (ICH). METHODS Altogether, 175 patients with ICH and 80 controls were encompassed in this prospective cohort study. Serum PINK1 levels were measured at admission of all patients and at study entry of all controls. The National Institutes of Health Stroke Scale (NIHSS) scores and hematoma volumes were applied to determine the severity. SAP, END and post-ICH 6-month poor prognosis (modified Rankin Scale scores: 3-6) were recorded as the three outcome variables of interest. RESULTS Patients, in contrast to controls, had significantly elevated serum PINK1 levels. Serum PINK1 levels were independently correlated with NIHSS scores, hematoma volumes and 6-month modified Rankin Scale scores. Serum PINK1 levels were linearly correlated with risks of SAP, END and post-ICH 6-month poor prognosis under the restricted cubic spline, as well as along with NIHSS scores and hematoma volumes, became their independent predictors. As demonstrated under receiver operating characteristic curve, serum PINK1 levels displayed effective predictive ability and possessed similar discrimination efficiency, when compared to NIHSS scores and hematoma volumes. Using sensitivity analysis, prognosis association was robust. CONCLUSION Serum PINK1 levels are substantially heightened after ICH, and may accurately mirror hemorrhagic intensity and efficaciously forecast END, SAP, and poor neurological prognosis, signifying that PINK1 may be a serological prognosticator of good prospect in ICH.
Collapse
Affiliation(s)
- Weifang Ni
- Department of Neurosurgery, The Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), No. 999 Zhongxing South Road, Yuecheng District, Shaoxing City 312000 Zhejiang Province, China
| | - Si-Hua Chen
- Department of Neurosurgery, The Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), No. 999 Zhongxing South Road, Yuecheng District, Shaoxing City 312000 Zhejiang Province, China.
| | - Le Dai
- Department of Neurosurgery, The Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), No. 999 Zhongxing South Road, Yuecheng District, Shaoxing City 312000 Zhejiang Province, China
| | - Yifu Zhou
- Department of Neurosurgery, The Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), No. 999 Zhongxing South Road, Yuecheng District, Shaoxing City 312000 Zhejiang Province, China
| | - Chenjun He
- Department of Neurosurgery, The Affiliated Hospital of Shaoxing University (Shaoxing Municipal Hospital), No. 999 Zhongxing South Road, Yuecheng District, Shaoxing City 312000 Zhejiang Province, China
| |
Collapse
|
2
|
Sun D, Gou H, Zhang Y, Li J, Dai C, Shen H, Chen K, Wang Y, Pan P, Zhu T, Xu C, Shan T, Liao M, Zhang J. Salmonella Typhimurium persistently infects host via its effector SseJ-induced PHB2-mediated mitophagy. Autophagy 2025; 21:1228-1244. [PMID: 39902787 DOI: 10.1080/15548627.2025.2462511] [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: 04/27/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
Despite decades of research on effective methods to resist Salmonella enterica serovar Typhimurium (S. Typhimurium) pathogenicity, the mechanisms of S. Typhimurium-host interactions have not been fully determined. S. Typhimurium is characterized as an important zoonosis in public health worldwide because of its endemicity, high morbidity, and difficulty in applying control and prevention measures. Herein, we introduce a novel bacterial factor, secretion system effector J (SseJ), and its interactive host protein, PHB2 (prohibitin 2). We explored whether SseJ affected S. Typhimurium replication and survival in the host. S. Typhimurium infection caused severe mitochondrial damage and mitophagy, which facilitated S. Typhimurium proliferation in cells. S. Typhimurium SseJ activated the PINK1 (PTEN induced kinase 1)-PRKN (parkin RBR E3 ubiquitin protein ligase)-autophagosome-dependent mitophagy pathway, aided by the mitophagy receptor PHB2, for bacterial survival and persistent infection. Moreover, suppression of mitophagy alleviated the pathogenicity of S. Typhimurium. In conclusion, S. Typhimurium infection could be antagonized by targeting the SseJ-PHB2-mediated host mitochondrial autophagy pathway.Abbreviation: ACTB: actin beta; BafA1: bafilomycin A1; CCCP: carbonyl cyanide m-chlorophenyl hydrazone; co-IP: co-immunoprecipitation; CFU: colony-forming units; COX4/COXIV: cytochrome c oxidase subunit 4; CQ: chloroquine; hpi: h post-bacterial infection; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; Mdivi-1:mitophagy inhibitor mitochondrial division inhibitor 1; MFN2: mitofusin 2; MG132: z-leu-leu-leucinal; MOI: multiplicity of infection; mtDNA: mitochondrial DNA; PBS: phosphate-buffered saline; PGAM5: PGAM family member 5, mitochondrial serine/threonine protein phosphatase; PHB2: prohibitin 2; PINK1: PTEN induced kinase 1; qPCR: quantitative real-time reverse transcription PCR; Roc-A: Rocaglamide A; PRKN/Parkin: parkin RBR E3 ubiquitin protein ligase; SCVs: Salmonella-containing vacuoles; siRNA: small interfering RNA; SPI-2: Salmonella pathogenicity island 2; SseJ: secretion system effector J; S. Typhimurium: Salmonella enterica serovar Typhimurium; S.T-ΔSseJ: SseJ gene-deleted Salmonella Typhimurium strains; S.T-CΔSseJ: SseJ-complemented Salmonella Typhimurium strains; WT: wild-type.
Collapse
Affiliation(s)
- Dage Sun
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongchao Gou
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Yu Zhang
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai, China
| | - Jiayi Li
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Changzhi Dai
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Haiyan Shen
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangdong Provincial Key Laboratory of Livestock Disease Prevention, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Guangzhou, China
| | - Kaifeng Chen
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yu Wang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Peng Pan
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ting Zhu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chenggang Xu
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Tongling Shan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Ming Liao
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Department of Preventive Veterinary Medicine, College of Animal Science and Technology, Zhongkai University of Agricultural Engineering, Guangzhou, China
| | - Jianmin Zhang
- National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Key Laboratory of Zoonoses, Ministry of Agriculture, Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
3
|
Oettinger D, Yamamoto A. Autophagy dysfunction and neurodegeneration: Where does it go wrong? J Mol Biol 2025:169219. [PMID: 40383464 DOI: 10.1016/j.jmb.2025.169219] [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: 01/30/2025] [Revised: 04/24/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
An infamous hallmark of neurodegenerative diseases is the accumulation of misfolded or unfolded proteins forming inclusions in the brain. The accumulation of these abnormal structures is a mysterious one, given that cells devote significant resources to integrate complementary pathways to ensure proteome integrity and proper protein folding. Aberrantly folded protein species are rapidly targeted for disposal by the ubiquitin-proteasome system (UPS), and even if this should fail, and the species accumulates, the cell can also rely on the lysosome-mediated degradation pathways of autophagy. Despite the many safeguards in place, failure to maintain protein homeostasis commonly occurs during, or preceding, the onset of disease. Over the last decade and a half, studies suggest that the failure of autophagy may explain the disruption in protein homeostasis observed in disease. In this review, we will examine how the highly complex cells of the brain can become vulnerable to failure of aggregate clearance at specific points during the processive pathway of autophagy, contributing to aggregate accumulation in brains with neurodegenerative disease.
Collapse
Affiliation(s)
- Daphne Oettinger
- Doctoral Program for Neurobiology and Behavior, Columbia University, New York, NY, USA
| | - Ai Yamamoto
- Departments of Neurology and Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
4
|
Singh PK, Agarwal S, Volpi I, Wilhelm LP, Becchi G, Keenlyside A, Macartney T, Toth R, Rousseau A, Masson GR, Ganley IG, Muqit MMK. Kinome screening identifies integrated stress response kinase EIF2AK1/HRI as a negative regulator of PINK1 mitophagy signaling. SCIENCE ADVANCES 2025; 11:eadn2528. [PMID: 40344059 PMCID: PMC12063660 DOI: 10.1126/sciadv.adn2528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Loss-of-function mutations in the PINK1 kinase lead to early-onset Parkinson's disease (PD). PINK1 is activated by mitochondrial damage to phosphorylate ubiquitin and Parkin, triggering mitophagy. PINK1 also indirectly phosphorylates Rab GTPases, such as Rab8A. Using an siRNA library targeting human Ser/Thr kinases in HeLa cells, we identified EIF2AK1 [heme-regulated inhibitor (HRI) kinase], a branch of the integrated stress response (ISR), as a negative regulator of PINK1. EIF2AK1 knockdown enhances mitochondrial depolarization-induced PINK1 stabilization and phosphorylation of ubiquitin and Rab8A. These results were confirmed in SK-OV-3, U2OS, and ARPE-19 cells. Knockdown of DELE1, an activator of EIF2AK1, produced similar effects. Notably, the ISR inhibitor ISRIB also enhanced PINK1 activation. In human cells with mito-QC mitophagy reporters, EIF2AK1 knockdown or ISRIB treatment increased PINK1-dependent mitophagy without affecting deferiprone-induced mitophagy. These findings suggest that the DELE1-EIF2AK1 ISR pathway is a negative regulator of PINK1-dependent mitophagy. Further evaluation in PD-relevant models is needed to assess the therapeutic potential of targeting this pathway.
Collapse
Affiliation(s)
- Pawan K. Singh
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Shalini Agarwal
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Ilaria Volpi
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Léa P. Wilhelm
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Giada Becchi
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Andrew Keenlyside
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Thomas Macartney
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Rachel Toth
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Adrien Rousseau
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Glenn R. Masson
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Ian G. Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Miratul M. K. Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| |
Collapse
|
5
|
Lee-Glover LP, Picard M, Shutt TE. Mitochondria - the CEO of the cell. J Cell Sci 2025; 138:jcs263403. [PMID: 40310473 PMCID: PMC12070065 DOI: 10.1242/jcs.263403] [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] [Indexed: 05/02/2025] Open
Abstract
As we have learned more about mitochondria over the past decades, including about their essential cellular roles and how altered mitochondrial biology results in disease, it has become apparent that they are not just powerplants pumping out ATP at the whim of the cell. Rather, mitochondria are dynamic information and energy processors that play crucial roles in directing dozens of cellular processes and behaviors. They provide instructions to enact programs that regulate various cellular operations, such as complex metabolic networks, signaling and innate immunity, and even control cell fate, dictating when cells should divide, differentiate or die. To help current and future generations of cell biologists incorporate the dynamic, multifaceted nature of mitochondria and assimilate modern discoveries into their scientific framework, mitochondria need a 21st century 'rebranding'. In this Opinion article, we argue that mitochondria should be considered as the 'Chief Executive Organelle' - the CEO - of the cell.
Collapse
Affiliation(s)
- Laurie P. Lee-Glover
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, 10032, USA
- Department of Neurology, H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia University Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, 10032, USA
- New York State Psychiatric Institute, New York, 10032, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Timothy E. Shutt
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
6
|
Lee CM, Chien TCR, Wang JS, Chen YW, Chen CY, Kuo CC, Chiang LT, Wu KK, Hsu WT. 5-Methoxytryptophan attenuates oxidative stress-induced downregulation of PINK1 and mitigates mitochondrial damage and apoptosis in cardiac myocytes. Free Radic Biol Med 2025; 232:398-411. [PMID: 40074188 DOI: 10.1016/j.freeradbiomed.2025.03.010] [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: 01/23/2025] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/14/2025]
Abstract
Mitochondrial dysfunction is a hallmark of the pathogenesis of various cardiovascular diseases. 5-Methoxytryptophan (5-MTP), an intrinsic amino acid metabolite, exerts cardioprotective effects potentially through the preservation of mitochondrial integrity. This study investigates the mechanisms and contexts in which 5-MTP positively impacts mitochondrial function using cultured human cardiac myocyte cells and HL-1 cardiac cells subjected to oxidative stress (OS). We first demonstrated that 5-MTP up-regulates the expression of PINK1, a key regulator of mitochondrial homeostasis. PINK1 knockdown attenuated the beneficial effects of 5-MTP on cardiomyocyte apoptosis. Furthermore, in cells exposed to OS, 5-MTP pretreatment led to a notable decrease in mitochondrial superoxide generation. Fluorescence imaging and network analysis showed that 5-MTP preserved mitochondrial membrane potential and enhanced mitochondrial network integrity. Reduced phosphorylation of dynamin-related protein 1, which is involved in mitochondrial fission, uncovered the role of 5-MTP in maintaining mitochondrial dynamics. Notably, 5-MTP attenuated OS-induced mitophagy, as evidenced by reduced mitophagy detection dye fluorescence and lower mitochondrial Parkin levels, suggesting that mechanisms beyond the PINK1/Parkin pathway are involved. Restoration of AKT phosphorylation and reduced mitochondrial Bax localization further revealed an additional pathway contributing to mitochondrial protection. Moreover, 5-MTP attenuated pro-apoptotic Bax levels and enhanced PINK1 expression in a rat model of ischemic cardiomyopathy, corroborating its cardioprotective role. Collectively, these findings demonstrate that 5-MTP mitigates mitochondrial dysfunction through coordinated regulation of PINK1, AKT, and Bax, offering potential as a therapeutic agent to enhance cellular resilience in OS-driven mitochondrial damage.
Collapse
Affiliation(s)
- Chii-Ming Lee
- Department of Cardiovascular Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Tung-Chun Russell Chien
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Juo-Shan Wang
- Department of Cardiovascular Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yu-Wei Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Yu Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Liang-Ting Chiang
- Department of Cardiovascular Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Kenneth K Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan; College of Life Sciences, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Wan-Tseng Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
7
|
Kay N, Huang CY, Yu YC, Chen CC, Chang CC, Huang SJ. The Involvement of Mitochondrial Dysfunction during the Development of Adenomyosis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:861-874. [PMID: 40010668 DOI: 10.1016/j.ajpath.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/10/2024] [Accepted: 01/10/2025] [Indexed: 02/28/2025]
Abstract
The etiology of adenomyosis remains unclear. The association between epithelial-mesenchymal transition (EMT) and mitochondrial dysfunction is involved in fibrotic diseases. Adenomyosis is defined as the existence of endometrial glands and stroma in the myometrium with EMT and ultimate fibrosis. This study was designed to investigate the involvement of mitochondrial dysfunction in fibrotic adenomyosis. Mitochondrial integrity was examined in mouse and human adenomyotic tissues. Control and tamoxifen-treated mice were treated with 3-nitropropionic acid (a mitochondrial dysfunction inducer) and NG-nitro-L-arginine methyl ester (a mitochondrial dysfunction inhibitor), respectively, at postnatal day 21, followed by an evaluation of adenomyosis, EMT, and fibrosis as well as the expression of mitophagy, oxidative stress, and transforming growth factor-β1 (TGF-β1). The gene profiles of adenomyotic uteri were examined at postnatal day 42. Adenomyotic mice exhibited increased development of EMT and fibrosis. Adenomyotic tissues showed consistent mitochondrial destruction with increased fission, mitophagosomes, and lysosomes. Besides, mitophagy, oxidative stress, and TGF-β1 levels were consistently increased. The mitochondrial dysfunction, the development of mitophagy and fibrosis, and TGF-β1 expression were induced by 3-nitropropionic acid in control uteri. In contrast, NG-nitro-L-arginine methyl ester attenuated mitochondrial dysfunction, mitophagy, fibrosis, and TGF-β1 in adenomyotic uteri. Gene profiling demonstrated increased expression of mitochondrial dysfunction-related genes in adenomyotic uteri. This indicates that mitochondrial dysfunction-induced TGF-β1 dysregulation and fibrosis are associated with the development of adenomyosis.
Collapse
Affiliation(s)
- Nari Kay
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chun-Yen Huang
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Ya-Chun Yu
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Chen Chen
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chi-Chang Chang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan
| | - S Joseph Huang
- Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, Morsani College of Medicine, University of South Florida, Tampa, Florida.
| |
Collapse
|
8
|
Yuan Y, Wang X, Cui Y, Zhou H, Li W, Teng Q, Wang H, Sun B, Wang Q, Sun H, Tang J. Attenuated PINK1 autophosphorylation play neuroprotective and anti-seizure roles in neonatal hypoxia. Sci Rep 2025; 15:15078. [PMID: 40301645 PMCID: PMC12041355 DOI: 10.1038/s41598-025-99915-8] [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] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
This study investigated the roles and mechanisms of PINK1 activity in neonatal hypoxia-induced seizures with shRNA intervention targeting translocase outer mitochondrial membrane 7 (TOM7), the positive regulator of PINK1 autophosphorylation, or overlapping with the m-AAA protease 1 homolog (OMA1), the negative regulator of PINK1 autophosphorylation. Studies have suggested that in hypoxia-induced neonatal seizures, the phosphorylation level of PINK1 is significantly increased and the mitophagic pathway is activated, accompanied by neuronal damage and learning-memory deficits. Inhibiting PINK1 phosphorylation by reducing TOM7 expression alleviated mitophagy, mitochondrial oxidative stress, neuronal damage and seizures. In contrast, the inhibition of OMA1 expression resulted in a further increase in PINK1 phosphorylation and aggravated hypoxia-induced seizures and neuronal injury. This study implicated PINK1 activity in neonatal hypoxia and suggest that attenuated PINK1 autophosphorylation may have neuroprotective and anti-seizure effects in neonatal hypoxia.
Collapse
Affiliation(s)
- Yi Yuan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xiaoqian Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yaru Cui
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Hua Zhou
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Wenna Li
- Affiliated Yantai Mountain Hospital, Binzhou Medical University, Yantai, 264003, China
| | - Qian Teng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Hongjin Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Bohan Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qiaoyun Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Jianhua Tang
- Affiliated Yantai Mountain Hospital, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
9
|
Chen H, Lin Q, Zeng Y, Chen P, Guo P, Feng R, Guo Z, Kang J, Chen Q, Zhou X. Xinyin tablets affect mitophagy and cardiomyocyte apoptosis to alleviate chronic heart failure by regulating histone deacetylase 3(HDAC3)-mediated PTEN induced putative kinase 1(PINK1)/Parkin signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119666. [PMID: 40122315 DOI: 10.1016/j.jep.2025.119666] [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: 10/10/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xinyin tablets, Chinese patent medicine, are composed of Panax ginseng C.A.Mey. (Araliaceae), Ilex pubescens Hook. & Arn. (Aquifoliaceae), Leonurus japonicus Houtt. (Lamiaceae), Plantago asiatica L. (Plantaginaceae), Ophiopogon japonicus (Thunb.) Ker Gawl. (Asparagaceae), Astragalus membranaceus (Fisch.) Bunge, and Draba nemorosa L. (Brassicaceae). It has been used for the prevention and treatment of chronic heart failure (CHF) clinically. However, its underlying mechanism of action is far from completely understood. AIM OF THE STUDY This study aimed to determine whether Xinyin tablets alleviate CHF in SPF C57 mice and to explore the potential mechanism of action in H9c2 cells. MATERIALS AND METHODS Liquid chromatography tandem mass spectroscopy (LC-MS/MS) was performed to identify the chemical compounds in Xinyin tablets. In vivo, 60 C57 mice were randomly divided into 6 groups: the sham group; model group; low-, medium-, and high-dose Xinyin tablets groups; and perindopril group. Animals in the sham group underwent thoracotomy only. The others were subjected to coronary artery ligation. After 4 weeks of drug intervention, the cardiac function of the mice in each group was detected via echocardiography, the myocardial cells were evaluated via HE staining, and the degree of myocardial fibrosis was detected via Masson's trichrome staining. The expression of PINK1/Parkin signaling pathway-related genes (HDAC3, PINK1, Parkin, P62, LC3II/I, caspase-3, caspase-9, and Bax) was analyzed via RT‒qPCR and Western blotting. The effects of Xinyin tablets on cardiomyocyte apoptosis and mitophagy mediated by the HDAC3 and PINK1/Parkin pathways in CHF model mice were evaluated. In vitro, H9c2 cardiomyocytes subjected to hypoxia were treated with different concentrations of Xinyin tablets. The mRNA transcription levels of HDAC3, PINK1, Parkin, P62, LC3II/I, caspase-3, caspase-9, and Bax were measured via fluorescence quantitative PCR. Western blotting was used to detect the protein expression levels of PINK1, Parkin, P62, LC3 II/I, caspase-3, caspase-9, and Bax. TUNEL staining was used to detect the number of apoptotic bodies in the myocardium to evaluate the level of apoptosis. Transmission electron microscopy was used to observe changes in the number of mitophagosomes. Rapamycin (mitophagy agonist), Mdivi-1 (mitophagy inhibitor), ITSA-1 (HDAC3 agonist) and RGFP966 (HDAC3 inhibitor) were used to create intervention conditions. The effects of rapamycin or Mdivi-1 on PINK1/Parkin-mediated mitophagy were observed. Then, the effects of HDAC3 on the PINK1/Parkin signaling pathway, mitophagy and apoptosis in hypoxic cardiomyocytes were observed. Hypoxic cardiomyocytes were treated with Xinyin tablets-containing serum or control serum to observe whether Xinyin tablets could still play a protective role in cardiomyocytes when HDAC3 is activated or mitophagy is inhibited. RESULTS 785 compounds were characterized from Xinyin tablets, among which carbohydrates and glycosides, phenylpropanoids, terpenes were abundant, and a small number of amino acids, peptides and derivatives also existed in Xinyin tablets. In vivo, Xinyin tablets improved cardiac function (LVEF, LVFS, LVEDD, LVESD, and LVESV) and downregulated the expression of caspase-3, caspase-9, and Bax. The expression levels of PINK1 and Parkin subsequently increased. In vitro, the above findings were reinforced in H9c2 cardiomyocytes. Rapamycin and RGFP966 reduced the apoptosis of hypoxic H9C2 cardiomyocytes and increased mitophagy mediated by the HDAC3-mediated PINK1/Parkin signaling pathway. CONCLUSIONS Xinyin tablets have potential as an intervention for CHF by improving mitophagy and inhibiting cardiomyocyte apoptosis through the HDAC3-mediated PINK1/Parkin signaling pathway.
Collapse
Affiliation(s)
- Hanyu Chen
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, China
| | - Qianbei Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanlin Zeng
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pinliang Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Pengpeng Guo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruoshui Feng
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhenyu Guo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinhua Kang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiucen Chen
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, China
| | - Xiaoxiong Zhou
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Clinical Research Academy of Chinese Medicine, China.
| |
Collapse
|
10
|
Valenti D, Abbrescia DI, Marzano F, Ravagnan G, Tullo A, Vacca RA. Polydatin reactivates mitochondrial bioenergetics and mitophagy while preventing premature senescence by modulating microRNA-155 and its direct targets in human fibroblasts with trisomy 21. Free Radic Biol Med 2025; 235:200-212. [PMID: 40280316 DOI: 10.1016/j.freeradbiomed.2025.04.032] [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: 01/29/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Mitochondrial dysfunction and redox dyshomeostasis are considered crucial factors causally linked to the pathogenesis of Down syndrome (DS), a human genetic anomaly currently lacking a cure, associated with neurodevelopmental deficits in children and early onset symptoms of aging in adults. Several natural plant-derived polyphenolic compounds, known for their neurostimulator, antioxidant and anti-inflammatory activities, have been proposed as dietary supplements to manage DS-linked phenotypic alterations. However, the poor bioavailability and rapid metabolism of these compounds have limited conclusive evidence regarding their clinical efficacy in individuals with DS. Polydatin (PLD), a natural polyphenolic glucoside precursor of resveratrol derived from Polygonum cuspidatum, is instead highly bioavailable and resistant to enzymatic oxidation. PLD supplementation has shown many therapeutic efficacies in several human diseases without side effects. In this study, we used fetal trisomy 21 human skin fibroblasts (DS-HSFs) to investigate, from a mechanistic point of view, whether PLD supplementation could prevent or counteract critical cellular alterations linked to both neurodevelopmental deficits and early aging in DS. Our findings demonstrate that PLD reactivates mitochondrial bioenergetics, reduces oxygen radical overproduction and prevents oxidative stress (OS)-induced cellular senescence and DNA damage in DS-HSF. Notably, we identified a novel mechanism of PLD action involving the chromosome-21-encoded microRNA-155 (miR-155) and its direct target genes casitas B-lineage lymphoma (CBL), BAG Cochaperone 5 (BAG5) and mitochondrial transcription factor A (TFAM). These proteins play pivotal roles in regulating mitochondrial bioenergetics, biogenesis and mitophagy. Given that the deregulation of miR-155/CBL axis is also implicated in acute leukemias, which frequently occur in children with DS, PLD emerges as a promising candidate for translational application. Its ability to enhance mitochondrial bioenergetics and address critical DS-associated phenotypic alterations highlights its therapeutic potential.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council of Italy (IBIOM-CNR), Via Amendola 122/O, 70126, Bari, Italy.
| | - Daniela Isabel Abbrescia
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council of Italy (IBIOM-CNR), Via Amendola 122/O, 70126, Bari, Italy
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council of Italy (IBIOM-CNR), Via Amendola 122/O, 70126, Bari, Italy
| | - Giampietro Ravagnan
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133, Rome, Italy
| | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council of Italy (IBIOM-CNR), Via Amendola 122/O, 70126, Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council of Italy (IBIOM-CNR), Via Amendola 122/O, 70126, Bari, Italy.
| |
Collapse
|
11
|
Basak B, Holzbaur ELF. Mitophagy in Neurons: Mechanisms Regulating Mitochondrial Turnover and Neuronal Homeostasis. J Mol Biol 2025:169161. [PMID: 40268233 DOI: 10.1016/j.jmb.2025.169161] [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: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Mitochondrial quality control is instrumental in regulating neuronal health and survival. The receptor-mediated clearance of damaged mitochondria by autophagy, known as mitophagy, plays a key role in controlling mitochondrial homeostasis. Mutations in genes that regulate mitophagy are causative for familial forms of neurological disorders including Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). PINK1/Parkin-dependent mitophagy is the best studied mitophagy pathway, while more recent work has brought to light additional mitochondrial quality control mechanisms that operate either in parallel to or independent of PINK1/Parkin mitophagy. Here, we discuss our current understanding of mitophagy mechanisms operating in neurons to govern mitochondrial homeostasis. We also summarize progress in our understanding of the links between mitophagic dysfunction and neurodegeneration, and highlight the potential for therapeutic interventions to maintain mitochondrial health and neuronal function.
Collapse
Affiliation(s)
- Bishal Basak
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
12
|
Feng Z, Hou Y, Yu C, Li T, Fu H, Lv F, Li P. Mitophagy in perioperative neurocognitive disorder: mechanisms and therapeutic strategies. Eur J Med Res 2025; 30:270. [PMID: 40211418 PMCID: PMC11987364 DOI: 10.1186/s40001-025-02400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/20/2025] [Indexed: 04/13/2025] Open
Abstract
Perioperative neurocognitive disorder (PND) is a common neurological complication after surgery/anesthesia in elderly patients that affect postoperative outcome and long-term quality of life, which increases the cost of family and social resources. The pathological mechanism of PND is complex and not fully understood, and the methods of prevention and treatment of PND are very limited, so it is particularly important to analyze the mechanism of PND. Research indicates that mitochondrial dysfunction is pivotal in the initiation and progression of PND, although the precise mechanisms remain elusive and could involve disrupted mitophagy. We reviewed recent studies on the link between mitophagy and PND, highlighting the role of key proteins in abnormal mitophagy and discussing therapeutic strategies aimed at mitophagy regulation. This provides insights into the mechanisms underlying PND and potential therapeutic targets.
Collapse
Affiliation(s)
- Zhen Feng
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, 301 Nancheng Avenue, Nan'an District, Chongqing, Chongqing, 400000, People's Republic of China
| | - Yan Hou
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, 301 Nancheng Avenue, Nan'an District, Chongqing, Chongqing, 400000, People's Republic of China
| | - Chang Yu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Ting Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Haoyang Fu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China.
| | - Ping Li
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, 1 Yuanjiagang Youyi Road, Yuzhong District, Chongqing, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
13
|
Wang W, Li Z, Lyu C, Wang T, Han C, Cui S, Wang J, Xu R. Mechanism of a Novel Complex: Zinc Oxide Nanoparticles-Luteolin to Promote Ferroptosis in Human Acute Myeloid Leukemia Cells in Vitro. Int J Nanomedicine 2025; 20:4035-4050. [PMID: 40191047 PMCID: PMC11972579 DOI: 10.2147/ijn.s509007] [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: 12/15/2024] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Purpose Acute myeloid leukemia (AML) is a hematological malignancy. Zinc oxide nanoparticles (ZnO NPs) and Luteolin are commonly used to fight cancer. In this study, we synthesized a new complex: zinc oxide nanoparticles-luteolin (ZnONPs-Lut) and aimed to investigate its effects on cell death in the AML cell line (MOLM-13) in vitro and to elucidate the underlying mechanisms. Methods We assessed cell viability, quantified changes in gene expression using real-time quantitative PCR (qRT-PCR), and measured changes in ferrous (Fe2+) content, glutathione (GSH) content, malondialdehyde (MDA) content, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) levels following treatment with different concentrations of MOLM-13 cells with different concentrations of ZnONPs-Lut. Western blotting was used to detect the protein expression levels of ACSL4, GPX4, FTH1, and SLC7A11, while the cell morphology was observed by transmission electron microscopy (TEM). Meanwhile, the effect of Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, on the expression of the aforementioned ferroptosis-related proteins and cell morphology was evaluated. Results The results showed that ZnONPs-Lut was able to significantly inhibit the proliferation of MOLM-13 cells in a time- and dose-dependent manner. Additionally, it increased the concentrations of Fe2+ and MDA, reduced the expression levels of GSH and MMP, and induced ROS generation. Furthermore, it also enhanced the expression of ACSL4 protein while decreasing the expression of GPX4, FTH1, and SLC7A11 proteins. Notably, Fer-1 was able to significantly restrain the changes in protein levels and mitochondrial morphology damage induced by ZnONPs-Lut after its action on cells. Conclusion ZnONPs-Lut inhibits the proliferation of MOLM-13 cells, likely through promoting the cellular ferroptosis signaling pathway. These findings suggest that ZnONPs-Lut could be a potential therapeutic approach for AML.
Collapse
Affiliation(s)
- Wenhao Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Zonghong Li
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Chunyi Lyu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Teng Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Chen Han
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Siyuan Cui
- Department of Hematology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, People’s Republic of China
| | - Jinxin Wang
- Department of Hematology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, People’s Republic of China
| | - Ruirong Xu
- Department of Hematology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, People’s Republic of China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Hematology, Health Commission of Shandong Province, Jinan, People’s Republic of China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| |
Collapse
|
14
|
Antico O, Thompson PW, Hertz NT, Muqit MMK, Parton LE. Targeting mitophagy in neurodegenerative diseases. Nat Rev Drug Discov 2025; 24:276-299. [PMID: 39809929 DOI: 10.1038/s41573-024-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Mitochondrial dysfunction is a hallmark of idiopathic neurodegenerative diseases, including Parkinson disease, amyotrophic lateral sclerosis, Alzheimer disease and Huntington disease. Familial forms of Parkinson disease and amyotrophic lateral sclerosis are often characterized by mutations in genes associated with mitophagy deficits. Therefore, enhancing the mitophagy pathway may represent a novel therapeutic approach to targeting an underlying pathogenic cause of neurodegenerative diseases, with the potential to deliver neuroprotection and disease modification, which is an important unmet need. Accumulating genetic, molecular and preclinical model-based evidence now supports targeting mitophagy in neurodegenerative diseases. Despite clinical development challenges, small-molecule-based approaches for selective mitophagy enhancement - namely, USP30 inhibitors and PINK1 activators - are entering phase I clinical trials for the first time.
Collapse
Affiliation(s)
- Odetta Antico
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul W Thompson
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura E Parton
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
15
|
Vlaikou AM, Nussbaumer M, Iliou A, Papageorgiou MP, Komini C, Theodoridou D, Benaki D, Mikros E, Gikas E, Syrrou M, Filiou MD. Early Life Stress Induces Brain Mitochondrial Dynamics Changes and Sex-Specific Adverse Effects in Adulthood. J Neurosci Res 2025; 103:e70023. [PMID: 40195806 DOI: 10.1002/jnr.70023] [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: 04/04/2023] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 04/09/2025]
Abstract
Early life stress exposure exerts detrimental effects in adulthood and is a risk factor for psychiatric disorders. Studies addressing the molecular mechanisms of early life stress have primarily focused on hormones and stress circuits. However, little is known on how mitochondria and mitochondrial dynamics (i.e., the orchestration of mitochondrial fission, fusion, mitophagy, and biogenesis) modulate early life stress responses. Here, we used a maternal separation with early weaning (MSEW) paradigm to investigate the behavioral and molecular early life stress-elicited effects in male and female C57BL/6 mice in adulthood. We first applied a behavioral test battery to assess MSEW-driven, anxiety-related and stress-coping alterations. We then looked for MSEW-induced, mitochondria-centered changes in cingulate cortex, hippocampus and cerebellum, as well as in plasma by combining protein, mRNA, mitochondrial DNA copy number (mtDNAcn) and metabolomics analyses. We found that MSEW mice are more anxious, show decreased antioxidant capacity in the cingulate cortex and have higher mRNA levels of the fission regulator Fis1 and the mitophagy activator Pink1 in the hippocampus, indicating a shift towards mitochondrial degradation. Hippocampal mRNA level alterations of apoptotic markers further suggest an MSEW-driven activation of apoptosis accompanied by a dysregulation of purine catabolism in the cerebellum in MSEW mice. Sex-specific analysis revealed distinct MSEW-induced changes in male and female mice at the molecular level. Our work reveals a previously unexplored role of mitochondrial dynamics in regulating early life stress effects and highlights a mitochondria-centered dysregulation as a persistent outcome of early life stress in adulthood.
Collapse
Affiliation(s)
- Angeliki-Maria Vlaikou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Markus Nussbaumer
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Aikaterini Iliou
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Maria P Papageorgiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Chrysoula Komini
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
| | - Daniela Theodoridou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Dimitra Benaki
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Emmanuel Mikros
- Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Evangelos Gikas
- Section of Analytical Chemistry, Department of Chemistry, School of Science, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Maria Syrrou
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Michaela D Filiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (BRI-FORTH), Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina, University of Ioannina, Ioannina, Greece
| |
Collapse
|
16
|
Raza A, Raina J, Sahu SK, Wadhwa P. Genetic mutations in kinases: a comprehensive review on marketed inhibitors and unexplored targets in Parkinson's disease. Neurol Sci 2025; 46:1509-1524. [PMID: 39760821 DOI: 10.1007/s10072-024-07970-2] [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: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
This comprehensive review navigates the landscape of genetic mutations in kinases, offering a thorough examination of both marketed inhibitors and unexplored targets in the context of Parkinson's Disease (PD). Although existing treatments for PD primarily center on symptom management, progress in comprehending the molecular foundations of the disease has opened avenues for targeted therapeutic approaches. This review encompasses an in-depth analysis of four key kinases-PINK1, LRRK2, GAK, and PRKRA-revealing that LRRK2 has garnered the most attention with a plethora of marketed inhibitors. However, the study underscores notable gaps in the exploration of inhibitors for PINK1, GAK, and a complete absence for PRKRA. The observed scarcity of inhibitors for these kinases emphasizes a significant area of untapped potential in PD therapeutics. By drawing attention to these unexplored targets, the review highlights the urgent need for focused research and drug development efforts to diversify the therapeutic landscape, potentially providing novel interventions for halting or slowing the progression of PD.
Collapse
Affiliation(s)
- Amir Raza
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Jeevika Raina
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar- Grand Trunk Rd, Phagwara, Punjab, India.
| |
Collapse
|
17
|
He X, Tao Y, Song C, He J, Gong D, Yu W, Wang H, Yu J, Yang X. Novel Parkin agonists from Poria cocos against dyslipidemia. Fitoterapia 2025; 182:106469. [PMID: 40054701 DOI: 10.1016/j.fitote.2025.106469] [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: 10/11/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/30/2025]
Abstract
Parkin, a cytosolic E3 ubiquitin ligase, plays a crucial role in targeting damaged mitochondria. The dysfunction of Parkin has been implicated in various diseases, including dyslipidemia, highlighting the significance of regulating Parkin activity for therapeutic interventions. Poria cocos (PC), a traditional Chinese medicine with a history spanning over two thousand years, has shown promising effects in regulating dyslipidemia. However, the scarcity of Parkin ligands, particularly from PC, remains a significant drawback in the field. This study identified two novel Parkin ligands from PC using a Parkin-based centrifugal ultrafiltration/liquid chromatography/mass spectrometry method. Molecular docking analysis, molecular dynamic simulations, and autoubiquitination assays confirmed their abilities to activate Parkin. Furthermore, their mitophagy promotion and dyslipidemia mitigation capacities were validated in fat emulsion-induced human liver L02 cells and high-fat diet-induced mice. The results revealed that the two ligands, tumulosic acid and polyporenic acid C, from PC activated Parkin and further promoted mitophagy to alleviate dyslipidemia. These findings will contribute to developing new drugs and enhance our understanding of the PC anti-dyslipidemia mechanisms.
Collapse
Affiliation(s)
- Xudong He
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China
| | - Yuxuan Tao
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China
| | - Chengzhu Song
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China
| | - Jinbiao He
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China; Southwest United Graduate School, 298 121st Street, Kunming 650092, China
| | - Dihong Gong
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China
| | - Weimei Yu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China
| | - Hui Wang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China; Hunan University of Chinese Medicine, 300 xueshi Road, Changsha 410208, China
| | - Jie Yu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China.
| | - Xingxin Yang
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, 1076 Yuhua Road, Kunming 650500, China; Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, 1076 Yuhua Road, Kunming 650500, China.
| |
Collapse
|
18
|
Rose K, Herrmann E, Kakudji E, Lizarrondo J, Celebi AY, Wilfling F, Lewis SC, Hurley JH. In situ cryo-ET visualization of mitochondrial depolarization and mitophagic engulfment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645001. [PMID: 40196634 PMCID: PMC11974748 DOI: 10.1101/2025.03.24.645001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Defective mitochondrial quality control in response to loss of mitochondrial membrane polarization is implicated in Parkinson's disease by mutations in PINK1 and PRKN. Application of in situ cryo-electron tomography (cryo-ET) made it possible to visualize the consequences of mitochondrial depolarization at higher resolution than heretofore attainable. Parkin-expressing U2OS cells were treated with the depolarizing agents oligomycin and antimycin A (OA), subjected to cryo-FIB milling, and mitochondrial structure was characterized by in situ cryo-ET. Phagophores were visualized in association with mitochondrial fragments. Bridge-like lipid transporter (BLTP) densities potentially corresponding to ATG2A were seen connected to mitophagic phagophores. Mitochondria in OA-treated cells were fragmented and devoid of matrix calcium phosphate crystals. The intermembrane gap of cristae was narrowed and the intermembrane volume reduced, and some fragments were devoid of cristae. A subpopulation of ATP synthases re-localized from cristae to the inner boundary membrane (IBM) apposed to the outer membrane (OMM). The structure of the dome-shaped prohibitin complex, a dodecamer of PHB1-PHB2 dimers, was determined in situ by sub-tomogram averaging in untreated and treated cells and found to exist in open and closed conformations, with the closed conformation is enriched by OA treatment. These findings provide a set of native snapshots of the manifold nano-structural consequences of mitochondrial depolarization and provide a baseline for future in situ dissection of Parkin-dependent mitophagy.
Collapse
Affiliation(s)
- Kevin Rose
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Eric Herrmann
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Eve Kakudji
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Javier Lizarrondo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - A Yasemin Celebi
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Florian Wilfling
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Samantha C Lewis
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - James H Hurley
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
19
|
Zanfardino P, Amati A, Perrone M, Petruzzella V. The Balance of MFN2 and OPA1 in Mitochondrial Dynamics, Cellular Homeostasis, and Disease. Biomolecules 2025; 15:433. [PMID: 40149969 PMCID: PMC11940761 DOI: 10.3390/biom15030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial dynamics, governed by fusion and fission, are crucial for maintaining cellular homeostasis, energy production, and stress adaptation. MFN2 and OPA1, key regulators of mitochondrial fusion, play essential roles beyond their structural functions, influencing bioenergetics, intracellular signaling, and quality control mechanisms such as mitophagy. Disruptions in these processes, often caused by MFN2 or OPA1 mutations, are linked to neurodegenerative diseases like Charcot-Marie-Tooth disease type 2A (CMT2A) and autosomal dominant optic atrophy (ADOA). This review explores the molecular mechanisms underlying mitochondrial fusion, the impact of MFN2 and OPA1 dysfunction on oxidative phosphorylation and autophagy, and their role in disease progression. Additionally, we discuss the divergent cellular responses to MFN2 and OPA1 mutations, particularly in terms of proliferation, senescence, and metabolic signaling. Finally, we highlight emerging therapeutic strategies to restore mitochondrial integrity, including mTOR modulation and autophagy-targeted approaches, with potential implications for neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neurosciences (DiBraiN), University of Bari Aldo Moro, Piazza Giulio Cesare, 70124 Bari, Italy; (P.Z.); (A.A.); (M.P.)
| |
Collapse
|
20
|
Mohanty S, Suklabaidya S, Mnatsakanyan N, Jacobson S, Harhaj EW. HTLV-1 Tax induces PINK1-Parkin-dependent mitophagy to mitigate activation of the cGAS-STING pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.15.643451. [PMID: 40161814 PMCID: PMC11952555 DOI: 10.1101/2025.03.15.643451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma (ATLL) and the neuroinflammatory disease, HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). The HTLV-1 Tax regulatory protein plays a critical role in HTLV-1 persistence and pathogenesis; however, the underlying mechanisms are poorly understood. Here we show that Tax dynamically regulates mitochondrial reactive oxygen species (ROS) and membrane potential to trigger mitochondrial dysfunction. Tax is recruited to damaged mitochondria through its interaction with the IKK regulatory subunit NEMO and directly engages the ubiquitin-dependent PINK1-Parkin pathway to induce mitophagy. Tax also recruits autophagy receptors NDP52 and p62/SQSTM1 to damaged mitochondria to induce mitophagy. Furthermore, Tax requires Parkin to limit the extent of cGAS-STING activation and suppress type I interferon (IFN). HTLV-1-transformed T cell lines and PBMCs from HAM/TSP patients exhibit hallmarks of chronic mitophagy which may contribute to immune evasion and pathogenesis. Collectively, our findings suggest that Tax manipulation of the PINK1-Parkin mitophagy pathway represents a new HTLV-1 immune evasion strategy.
Collapse
Affiliation(s)
- Suchitra Mohanty
- Department of Cell and Biological Systems, Penn State College School of Medicine, Hershey, PA 17033, USA
| | - Sujit Suklabaidya
- Department of Cell and Biological Systems, Penn State College School of Medicine, Hershey, PA 17033, USA
| | - Nelli Mnatsakanyan
- Department of Cell and Biological Systems, Penn State College School of Medicine, Hershey, PA 17033, USA
| | - Steven Jacobson
- Viral Immunology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Edward W. Harhaj
- Department of Cell and Biological Systems, Penn State College School of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
21
|
Park NY, Jo DS, Park HJ, Bae JE, Kim YH, Kim JB, Lee HJ, Kim SH, Choi H, Lee HS, Yoshimori T, Lee DS, Lee JA, Kim P, Cho DH. Deciphering melanophagy: role of the PTK2-ITCH-MLANA-OPTN cascade on melanophagy in melanocytes. Autophagy 2025; 21:664-673. [PMID: 39477686 PMCID: PMC11849925 DOI: 10.1080/15548627.2024.2421695] [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: 01/16/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/13/2024] Open
Abstract
Melanosomes play a pivotal role in skin color and photoprotection. In contrast to the well-elucidated pathway of melanosome biogenesis, the process of melanosome degradation, referred to as melanophagy, is largely unexplored. Previously, we discovered that 3,4,5-trimethoxycinnamate thymol ester (TCTE) effectively inhibits skin pigmentation by activating melanophagy. In this study, we discovered a new regulatory signaling cascade that controls melanophagy in TCTE-treated melanocytes. ITCH (itchy E3 ubiquitin protein ligase) facilitates ubiquitination of the melanosome membrane protein MLANA (melan-A) during TCTE-induced melanophagy. This ubiquitinated MLANA is then recognized by an autophagy receptor protein, OPTN (optineurin). Additionally, a phospho-kinase antibody array revealed that TCTE activates PTK2 (protein tyrosine kinase 2), which phosphorylates ITCH, enhancing the ubiquitination of MLANA. Furthermore, inhibition of either PTK2 or ITCH disrupts the ubiquitination of MLANA and the MLANA-OPTN interaction in TCTE-treated cells. Taken together, our findings highlight the critical role of the PTK2-ITCH-MLANA-OPTN cascade in orchestrating melanophagy progression.Abbreviations: α-MSH: alpha-melanocyte-stimulating hormone; dichlone: 2,3-dichloro-1,4-naphthoquinone; ITCH: itchy E3 ubiquitin protein ligase; MITF: melanocyte inducing transcription factor; MLANA: melan-A; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PINK1: PTEN induced kinase 1; PTK2: protein tyrosine kinase 2; SQSTM1/p62: sequestosome 1; TCTE: 3,4,5-trimethoxycinnamate thymol ester; TPC2: two pore segment channel 2; VDAC1: voltage dependent anion channel 1.
Collapse
Affiliation(s)
- Na Yeon Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Doo Sin Jo
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Hyun Jun Park
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Ji-Eun Bae
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Hwan Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Joon Bum Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Ha Jung Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Sung Hyun Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Hyunjung Choi
- R&D Unit, AmorePacific Corporation, Yongin, Gyeonggi-Do, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Dong-Seok Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- Organelle Institute, KNU, Daegu, Republic of Korea
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Republic of Korea
| | - Pansoo Kim
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Dong-Hyung Cho
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
- Organelle Institute, KNU, Daegu, Republic of Korea
| |
Collapse
|
22
|
Ma Y, Erb ML, Moore DJ. Aging, cellular senescence and Parkinson's disease. JOURNAL OF PARKINSON'S DISEASE 2025; 15:239-254. [PMID: 39973488 DOI: 10.1177/1877718x251316552] [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: 02/21/2025]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative movement disorder, affecting 1-2% of people over age 65. The risk of developing PD dramatically increases with advanced age, indicating that aging is likely a driving factor in PD neuropathogenesis. Several age-associated biological changes are also hallmarks of PD neuropathology, including mitochondrial dysfunction, oxidative stress, and neuroinflammation. Accumulation of senescent cells is an important feature of aging that contributes to age-related diseases. How age-related cellular senescence affects brain health and whether this phenomenon contributes to neuropathogenesis in PD is not yet fully understood. In this review, we highlight hallmarks of aging, including mitochondrial dysfunction, loss of proteostasis, genomic instability and telomere attrition in relation to well established PD neuropathological pathways. We then discuss the hallmarks of cellular senescence in the context of neuroscience and review studies that directly examine cellular senescence in PD. Studying senescence in PD presents challenges and holds promise for advancing our understanding of disease mechanisms, which could contribute to the development of effective disease-modifying therapeutics. Targeting senescent cells or modulating the senescence-associated secretory phenotype (SASP) in PD requires a comprehensive understanding of the complex relationship between PD pathogenesis and cellular senescence.
Collapse
Affiliation(s)
- Yue Ma
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Madalynn L Erb
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Darren J Moore
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
23
|
Dong Q, Zhu Y, Zhang X, Li L, Yang Y, Liu C, Wen J. Phytochemicals Targeting Mitophagy to Treat Heart Diseases: Retrospective Insights and Prospective Directions. Phytother Res 2025; 39:1592-1614. [PMID: 39912509 DOI: 10.1002/ptr.8448] [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: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 02/07/2025]
Abstract
Mitophagy is a process by which cells selectively eliminate damaged or dysfunctional mitochondria through the autophagy-lysosome pathway, thereby maintaining mitochondrial quality and cellular homeostasis. This process is closely linked to the onset and progression of various heart diseases. Modern pharmacological research has demonstrated that phytochemicals can regulate mitochondrial homeostasis in cardiomyocytes through multiple mechanisms, influencing mitophagy and protecting cardiomyocytes, which in turn exerts anti-cardiovascular effects. However, the underlying mechanisms of these effects are not yet fully understood. This study summarizes the pharmacological effects and molecular mechanisms of mitophagy in heart diseases, aiming to provide reference for the research and treatment of phytochemicals targeting mitophagy against heart diseases. The results indicated that phytochemicals (such as Berberine, Ginsenoside Rg1, Quercetin, Resveratrol, Baicalein, and so on) can exert preventive and therapeutic effects on heart diseases (such as cardiac toxicity or damage, myocardial ischemia/reperfusion injury, heart failure, heart aging, cardiac hypertrophy, cardiomyopathy, and so on.) via regulating the PINK1/Parkin and FUNDC1-dependent mitophagy pathway. These compounds mainly exert their effects by regulating mitochondrial homeostasis, mitochondrial dynamics, mitochondrial oxidative stress, mitochondrial apoptosis, and mitochondrial energy metabolism. This study provides a reference that phytochemicals have effect on anti-cardiovascular effects by regulating mitophagy. However, further in-depth mechanistic and clinical research are needed in the future.
Collapse
Affiliation(s)
- Qin Dong
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yichan Zhu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Xinghai Zhang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Lu Li
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Yi Yang
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Chuan Liu
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| | - Jianxia Wen
- School of Food and Bioengineering, Food Microbiology Key Laboratory of Sichuan Province, Chongqing Key Laboratory of Sichuan Chongqing Joint Construction of Specialty Food, Xihua University, Chengdu, China
| |
Collapse
|
24
|
Jauhari A, Monek AC, Suofu Y, Amygdalos OR, Singh T, Baranov SV, Carlisle DL, Friedlander RM. Melatonin Deficits Result in Pathologic Metabolic Reprogramming in Differentiated Neurons. J Pineal Res 2025; 77:e70037. [PMID: 39982401 PMCID: PMC11844733 DOI: 10.1111/jpi.70037] [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: 09/15/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Differentiation from neural progenitor to mature neuron requires a metabolic switch, whereby mature neurons become almost entirely dependent upon oxidative phosphorylation (OXPHOS) for ATP production. Although more efficient with respect to ATP production, OXPHOS produces additional reactive oxygen species, as compared to glycolysis; thus, endogenous mechanisms to quench free radicals are essential for the maintenance of neuronal health. Melatonin is synthesized in neuronal mitochondria and has a dual role as a free radical scavenger and as an inhibitor of mitochondrial-triggered cell death and proinflammatory pathways. Previously, we showed that loss of endogenous melatonin induced mitochondrial DNA (mtDNA) and cytochrome c (CytC) release triggering pathological inflammation and cell death pathways, respectively. Here we find that in mature neurons, but not undifferentiated neuronal cells, melatonin deficiency altered metabolic reprogramming in aralkylamine N-acetyltransferase knockout (AANAT-KO) neurons as compared with neurons expressing AANAT. Interestingly, there are no differences in neural progenitors regardless of AANAT status. In addition, AANAT-KO deficiency elevated BAK and BAX levels in AANAT-KO neurons. Further, we found that exogenous melatonin treatment of AANAT-KO cells during differentiation into mature neurons rescued metabolic reprogramming defects and restored normal BAK/BAX levels. Thus, we demonstrated that the metabolic reprogramming and subsequent consequences of the switch to OXPHOS that normally occurs during neuronal maturation are compromised by melatonin deficiency and rescued by melatonin supplementation.
Collapse
Affiliation(s)
- Abhishek Jauhari
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Adam C. Monek
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Yalikun Suofu
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Olivia R. Amygdalos
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Tanisha Singh
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Sergei V. Baranov
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Diane L. Carlisle
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Robert M. Friedlander
- Neuroapoptosis Laboratory, Department of Neurological SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
25
|
Pollock L, Georgiou IC, Rusilowicz-Jones EV, Clague MJ, Urbé S. A long-lived pool of PINK1 imparts a molecular memory of depolarization-induced activity. SCIENCE ADVANCES 2025; 11:eadr1938. [PMID: 40020067 PMCID: PMC11870087 DOI: 10.1126/sciadv.adr1938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/24/2025] [Indexed: 03/03/2025]
Abstract
The Parkinson's disease-linked kinase, PINK1, is a short-lived protein that undergoes cleavage upon mitochondrial import leading to its proteasomal degradation. Under depolarizing conditions, it accumulates on mitochondria where it becomes activated, phosphorylating both ubiquitin and the ubiquitin E3 ligase Parkin, at Ser65. Our experiments reveal that in retinal pigment epithelial cells, only a fraction of PINK1 becomes stabilized after depolarization by electron transport chain inhibitors. Furthermore, the observed accrual of PINK1 cannot be completely accounted for without an accompanying increase in biosynthesis. We have used a ubiquitylation inhibitor TAK-243 to accumulate cleaved PINK1. Under these conditions, generation of unconjugated "free" phospho-ubiquitin serves as a proxy readout for PINK1 activity. This has enabled us to find a preconditioning phenomenon, whereby an initial depolarizing treatment leaves a residual pool of active PINK1 that remains competent to seed the activation of nascent cleaved PINK1 following a 16-hour recovery period.
Collapse
Affiliation(s)
- Liam Pollock
- Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 3BX, UK
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH-Alliance, 69120 Heidelberg, Germany
| | - Ioanna Ch. Georgiou
- Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Emma V. Rusilowicz-Jones
- Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Michael J. Clague
- Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Sylvie Urbé
- Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| |
Collapse
|
26
|
Huang W, Lou A, Wang J, Wang Y, Zhang W, Li J, Wang S, Geng S, Wang G, Li X. TMBIM1 ameliorates sepsis-induced cardiac dysfunction by promoting Parkin-mediated mitophagy. FASEB J 2025; 39:e70397. [PMID: 39937566 DOI: 10.1096/fj.202402599rr] [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: 10/28/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
Myocardial dysfunction is a significant complication of sepsis that is associated with elevated mortality rates. Transmembrane BAX inhibitor motif containing 1 (TMBIM1), a stress-responsive protein, has garnered interest in the field of cardiovascular disease for its cardioprotective properties. Nevertheless, the role of TMBIM1 on sepsis-induced cardiac dysfunction (SICD) remains unknown. Here, our findings revealed a significant elevation in TMBIM1 expression within the myocardium following endotoxin challenge and further demonstrate the cardioprotective effects of TMBIM1 through adenovirus-mediated gene manipulation. Notably, lipopolysaccharide exposure markedly induced mitochondrial dysfunction in cardiomyocytes, which was effectively alleviated by TMBIM1 overexpression, while TMBIM1 knockdown exacerbated this dysfunction. Moreover, in cardiomyocytes subjected to endotoxin challenge, TMBIM1 was observed to interact with Parkin, facilitating its translocation from the cytosol to damaged mitochondria. This interaction enhanced the activation of mitophagy, thereby promoting the clearance of dysfunctional mitochondria and subsequently mitigating cellular injury. Hence, targeting TMBIM1 could be a novel therapeutic strategy for treating SICD.
Collapse
Affiliation(s)
- Weichang Huang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Critical Care Medicine, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan, China
| | - Anni Lou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuegang Wang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenyong Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, Lishui, Zhejiang, China
| | - Shiyu Geng
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guozhen Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Yang P, Shuai W, Wang X, Hu X, Zhao M, Wang A, Wu Y, Ouyang L, Wang G. Mitophagy in Neurodegenerative Diseases: Mechanisms of Action and the Advances of Drug Discovery. J Med Chem 2025; 68:3970-3994. [PMID: 39908485 DOI: 10.1021/acs.jmedchem.4c01779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Neurodegenerative diseases (NDDs), such as Parkinson's disease (PD) and Alzheimer's disease (AD), are devastating brain diseases and are incurable at the moment. Increasing evidence indicates that NDDs are associated with mitochondrial dysfunction. Mitophagy removes defective or redundant mitochondria to maintain cell homeostasis, whereas deficient mitophagy accelerates the accumulation of damaged mitochondria to mediate the pathologies of NDDs. Therefore, targeting mitophagy has become a valuable therapeutic pathway for the treatment of NDDs. Several mitophagy modulators have been shown to ameliorate neurodegeneration in PD and AD. However, it remains to be further investigated for other NDDs. Here, we describe the mechanism and key signaling pathway of mitophagy and summarize the roles of defective mitophagy on the pathogenesis of NDDs. Further, we underline the development advances of mitophagy modulators for PD and AD therapy, discuss the therapeutic challenges and limitations of the existing modulators, and provide guidelines for mitophagy mechanism exploration and drug design.
Collapse
Affiliation(s)
- Panpan Yang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Wen Shuai
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xin Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Xiuying Hu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Aoxue Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yongya Wu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Ding Z, Li Z, Sun K, Liu Y, Fang Z, Sun S, Li C, Wang Z. Mitochondrial Regulation of Ferroptosis in Cancer Cells. Int J Biol Sci 2025; 21:2179-2200. [PMID: 40083691 PMCID: PMC11900798 DOI: 10.7150/ijbs.105446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Ferroptosis is an iron-dependent nonapoptotic regulated cell death modality characterized by lethal levels of lipid peroxide accumulation and disrupted antioxidant systems. An increasing number of studies have revealed correlations between ferroptosis and the pathophysiology and treatment of cancer. Given the intricate involvement of mitochondria in ferroptosis, as suggested by previous studies, here, we review advances in understanding the roles of mitochondrial quality control and mitochondrial metabolism (including the roles of the TCA cycle, reactive oxygen species, iron metabolism, and lipid metabolism) in cancer-related ferroptosis and outline the molecular mechanism and clinical translation of mitochondria-related ferroptosis in cancer treatment. with the aim of promoting the precise utilization and prevention of ferroptosis in cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Shengrong Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Chenyuan Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhong Wang
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
29
|
Quan T, Li R, Gao T. Role of Mitochondrial Dynamics in Skin Homeostasis: An Update. Int J Mol Sci 2025; 26:1803. [PMID: 40076431 PMCID: PMC11898645 DOI: 10.3390/ijms26051803] [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: 01/10/2025] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 03/14/2025] Open
Abstract
Skin aging is the most prominent phenotype of host aging and is the consequence of a combination of genes and environment. Improving skin aging is essential for maintaining the healthy physiological function of the skin and the mental health of the human body. Mitochondria are vital organelles that play important roles in cellular mechanisms, including energy production and free radical balance. However, mitochondrial metabolism, mitochondrial dynamics, biogenesis, and degradation processes vary greatly in various cells in the skin. It is well known that mitochondrial dysfunction can promote the aging and its associated diseases of the skin, resulting in the damage of skin physiology and the occurrence of skin pathology. In this review, we summarize the important role of mitochondria in various skin cells, review the cellular responses to vital steps in mitochondrial quality regulation, mitochondrial dynamics, mitochondrial biogenesis, and mitochondrial phagocytosis, and describe their importance and specific pathways in skin aging.
Collapse
Affiliation(s)
| | | | - Ting Gao
- College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (T.Q.); (R.L.)
| |
Collapse
|
30
|
Chen TH, Lin SH, Lee MY, Wang HC, Tsai KF, Chou CK. Mitochondrial alterations and signatures in hepatocellular carcinoma. Cancer Metastasis Rev 2025; 44:34. [PMID: 39966277 PMCID: PMC11836208 DOI: 10.1007/s10555-025-10251-9] [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: 08/06/2024] [Accepted: 02/09/2025] [Indexed: 02/20/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer worldwide. Its primary risk factors are chronic liver diseases such as metabolic fatty liver disease, non-alcoholic steatohepatitis, and hepatitis B and C viral infections. These conditions contribute to a specific microenvironment in liver tumors which affects mitochondrial function. Mitochondria are energy producers in cells and are responsible for maintaining normal functions by controlling mitochondrial redox homeostasis, metabolism, bioenergetics, and cell death pathways. HCC involves abnormal mitochondrial functions, such as accumulation of reactive oxygen species, oxidative stress, hypoxia, impairment of the mitochondrial unfolded protein response, irregularities in mitochondrial dynamic fusion/fission mechanisms, and mitophagy. Cell death mechanisms, such as necroptosis, pyroptosis, ferroptosis, and cuproptosis, contribute to hepatocarcinogenesis and play a significant role in chemoresistance. The relationship between mitochondrial dynamics and HCC is thus noteworthy. In this review, we summarize the recent advances in mitochondrial alterations and signatures in HCC and attempt to elucidate its molecular biology. Here, we provide an overview of the mitochondrial processes involved in hepatocarcinogenesis and offer new insights into the molecular pathology of the disease. This may help guide future research focused on improving patient outcomes using innovative therapies.
Collapse
Affiliation(s)
- Tsung-Hsien Chen
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan
| | - Shu-Hsien Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan
| | - Ming-Yang Lee
- Division of Hemato-Oncology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan
- Min-Hwei Junior College of Health Care Management, Tainan, 73658, Taiwan
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, Chiayi, 62102, Taiwan
| | - Kun-Feng Tsai
- Department of Internal Medicine, Gastroenterology and Hepatology Section, An Nan Hospital, China Medical University, Tainan, 70965, Taiwan.
- Department of Medical Sciences Industry, Chang Jung Christian University, Tainan, 71101, Taiwan.
| | - Chu-Kuang Chou
- Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
- Obesity Center, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
- Department of Medical Quality, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
| |
Collapse
|
31
|
Joaquim M, Altin S, Bulimaga MB, Simões T, Nolte H, Bader V, Franchino CA, Plouzennec S, Szczepanowska K, Marchesan E, Hofmann K, Krüger M, Ziviani E, Trifunovic A, Chevrollier A, Winklhofer KF, Motori E, Odenthal M, Escobar-Henriques M. Mitofusin 2 displays fusion-independent roles in proteostasis surveillance. Nat Commun 2025; 16:1501. [PMID: 39929801 PMCID: PMC11811173 DOI: 10.1038/s41467-025-56673-5] [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: 07/21/2023] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Mitochondria are essential organelles and their functional state dictates cellular proteostasis. However, little is known about the molecular gatekeepers involved, especially in absence of external stress. Here we identify a role of MFN2 in quality control independent of its function in organellar shape remodeling. MFN2 ablation alters the cellular proteome, marked for example by decreased levels of the import machinery and accumulation of the kinase PINK1. Moreover, MFN2 interacts with the proteasome and cytosolic chaperones, thereby preventing aggregation of newly translated proteins. Similarly to MFN2-KO cells, patient fibroblasts with MFN2-disease variants recapitulate excessive protein aggregation defects. Restoring MFN2 levels re-establishes proteostasis in MFN2-KO cells and rescues fusion defects of MFN1-KO cells. In contrast, MFN1 loss or mitochondrial shape alterations do not alter protein aggregation, consistent with a fusion-independent role of MFN2 in cellular homeostasis. In sum, our findings open new possibilities for therapeutic strategies by modulation of MFN2 levels.
Collapse
Affiliation(s)
- Mariana Joaquim
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Selver Altin
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Maria-Bianca Bulimaga
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Pathology, Medical Faculty of the University of Cologne and University Hospital of Cologne, Cologne, Germany
| | - Tânia Simões
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Hendrik Nolte
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- MPI for Biology of Ageing, 50931, Cologne, Germany
| | - Verian Bader
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany, and Cluster of Excellence RESOLV, Bochum, Germany
| | - Camilla Aurora Franchino
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Solenn Plouzennec
- University of Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Karolina Szczepanowska
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- ReMedy International Research Agenda Unit, International Institute of Molecular Mechanisms and Machines (IMol), Polish Academy of Sciences, 00-783, Warsaw, Poland
| | | | - Kay Hofmann
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Marcus Krüger
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elena Ziviani
- Deparment of Biology, University of Padova, Padova, Italy
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Arnaud Chevrollier
- University of Angers, MitoLab Team, MitoVasc Unit, CNRS UMR6015, INSERM U1083, SFR ICAT, Angers, France
| | - Konstanze F Winklhofer
- Department Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Ruhr University Bochum, Germany, and Cluster of Excellence RESOLV, Bochum, Germany
| | - Elisa Motori
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Pathology, Medical Faculty of the University of Cologne and University Hospital of Cologne, Cologne, Germany
| | - Mafalda Escobar-Henriques
- Institute for Genetics, University of Cologne, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
32
|
Eskander G, Abdelhamid SG, Wahdan SA, Radwan SM. Insights on the crosstalk among different cell death mechanisms. Cell Death Discov 2025; 11:56. [PMID: 39929794 PMCID: PMC11811070 DOI: 10.1038/s41420-025-02328-9] [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: 08/06/2024] [Revised: 12/23/2024] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
The phenomenon of cell death has garnered significant scientific attention in recent years, emerging as a pivotal area of research. Recently, novel modalities of cellular death and the intricate interplay between them have been unveiled, offering insights into the pathogenesis of various diseases. This comprehensive review delves into the intricate molecular mechanisms, inducers, and inhibitors of the underlying prevalent forms of cell death, including apoptosis, autophagy, ferroptosis, necroptosis, mitophagy, and pyroptosis. Moreover, it elucidates the crosstalk and interconnection among the key pathways or molecular entities associated with these pathways, thereby paving the way for the identification of novel therapeutic targets, disease management strategies, and drug repurposing.
Collapse
Affiliation(s)
- Georgette Eskander
- Postgraduate program, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | - Sara A Wahdan
- Pharmacology and toxicology Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Sara M Radwan
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
33
|
Borbolis F, Ploumi C, Palikaras K. Calcium-mediated regulation of mitophagy: implications in neurodegenerative diseases. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:4. [PMID: 39911695 PMCID: PMC11790495 DOI: 10.1038/s44324-025-00049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025]
Abstract
Calcium signaling plays a pivotal role in diverse cellular processes through precise spatiotemporal regulation and interaction with effector proteins across distinct subcellular compartments. Mitochondria, in particular, act as central hubs for calcium buffering, orchestrating energy production, redox balance and apoptotic signaling, among others. While controlled mitochondrial calcium uptake supports ATP synthesis and metabolic regulation, excessive accumulation can trigger oxidative stress, mitochondrial membrane permeabilization, and cell death. Emerging findings underscore the intricate interplay between calcium homeostasis and mitophagy, a selective type of autophagy for mitochondria elimination. Although the literature is still emerging, this review delves into the bidirectional relationship between calcium signaling and mitophagy pathways, providing compelling mechanistic insights. Furthermore, we discuss how disruptions in calcium homeostasis impair mitophagy, contributing to mitochondrial dysfunction and the pathogenesis of common neurodegenerative diseases.
Collapse
Affiliation(s)
- Fivos Borbolis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Ploumi
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
34
|
Cho YL, Tan HWS, Yang J, Kuah BZM, Lim NSY, Fu N, Bay BH, Ling SC, Shen HM. Glucose-6-phosphate dehydrogenase regulates mitophagy by maintaining PINK1 stability. LIFE METABOLISM 2025; 4:loae040. [PMID: 39872984 PMCID: PMC11749863 DOI: 10.1093/lifemeta/loae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme in the pentose phosphate pathway (PPP) in glycolysis. Glucose metabolism is closely implicated in the regulation of mitophagy, a selective form of autophagy for the degradation of damaged mitochondria. The PPP and its key enzymes such as G6PD possess important metabolic functions, including biosynthesis and maintenance of intracellular redox balance, while their implication in mitophagy is largely unknown. Here, via a whole-genome CRISPR-Cas9 screening, we identified that G6PD regulates PINK1 (phosphatase and tensin homolog [PTEN]-induced kinase 1)-Parkin-mediated mitophagy. The function of G6PD in mitophagy was verified via multiple approaches. G6PD deletion significantly inhibited mitophagy, which can be rescued by G6PD reconstitution. Intriguingly, while the catalytic activity of G6PD is required, the known PPP functions per se are not involved in mitophagy regulation. Importantly, we found a portion of G6PD localized at mitochondria where it interacts with PINK1. G6PD deletion resulted in an impairment in PINK1 stabilization and subsequent inhibition of ubiquitin phosphorylation, a key starting point of mitophagy. Finally, we found that G6PD deletion resulted in lower cell viability upon mitochondrial depolarization, indicating the physiological function of G6PD-mediated mitophagy in response to mitochondrial stress. In summary, our study reveals a novel role of G6PD as a key positive regulator in mitophagy, which bridges several important cellular processes, namely glucose metabolism, redox homeostasis, and mitochondrial quality control.
Collapse
Affiliation(s)
- Yik-Lam Cho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
| | - Hayden Weng Siong Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Jicheng Yang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Basil Zheng Mian Kuah
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Nicole Si Ying Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Naiyang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Programs in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117549, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Faculty of Health Sciences, MOE Frontier Centre for Precision Oncology, University of Macau, Macao 999078, China
| |
Collapse
|
35
|
Ghiglione N, Abbo D, Bushunova A, Costamagna A, Porporato PE, Martini M. Metabolic plasticity in pancreatic cancer: The mitochondrial connection. Mol Metab 2025; 92:102089. [PMID: 39736443 PMCID: PMC11846432 DOI: 10.1016/j.molmet.2024.102089] [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: 10/16/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Cellular metabolism plays a pivotal role in the development and progression of pancreatic ductal adenocarcinoma (PDAC), with dysregulated metabolic pathways contributing to tumorigenesis and therapeutic resistance. Distinct metabolic heterogeneity in pancreatic cancer significantly impacts patient prognosis, as variations in metabolic profiles influence tumor behavior and treatment responses. SCOPE OF THE REVIEW This review explores the intricate interplay between mitochondrial dynamics, mitophagy, and cellular metabolism in PDAC. We discuss the significance of mitophagy dysregulation in PDAC pathogenesis, emphasizing its influence on treatment responses and prognosis. Furthermore, we analyze the impact of mitochondrial dynamics alterations, including fission and fusion processes, on PDAC progression and tumorigenesis. MAJOR CONCLUSION Targeting mitochondrial metabolism holds promise for advancing PDAC therapeutics. Ongoing clinical trials underscore the therapeutic potential of modulating key regulators of mitochondrial dynamics and mitophagy. Despite inherent challenges, these approaches offer diverse strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Noemi Ghiglione
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) Guido Tarone, University of Turin, Torino, Italy
| | - Damiano Abbo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) Guido Tarone, University of Turin, Torino, Italy
| | - Anastasia Bushunova
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) Guido Tarone, University of Turin, Torino, Italy
| | - Andrea Costamagna
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) Guido Tarone, University of Turin, Torino, Italy
| | - Paolo Ettore Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) Guido Tarone, University of Turin, Torino, Italy
| | - Miriam Martini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center (MBC) Guido Tarone, University of Turin, Torino, Italy.
| |
Collapse
|
36
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-z] [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: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
37
|
Sisalli MJ, D'Apolito E, Cuomo O, Lombardi G, Tufano M, Annunziato L, Scorziello A. The E3-ligase Siah2 activates mitochondrial quality control in neurons to maintain energy metabolism during ischemic brain tolerance. Cell Death Dis 2025; 16:52. [PMID: 39875361 PMCID: PMC11775118 DOI: 10.1038/s41419-025-07339-z] [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: 11/11/2023] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025]
Abstract
Mitochondrial quality control is crucial for the homeostasis of the mitochondrial network. The balance between mitophagy and biogenesis is needed to reduce cerebral ischemia-induced cell death. Ischemic preconditioning (IPC) represents an adaptation mechanism of CNS that increases tolerance to lethal cerebral ischemia. It has been demonstrated that hypoxia-induced Seven in absentia Homolog 2 (Siah2) E3-ligase activation influences mitochondrial dynamics promoting the degradation of mitochondrial proteins. Therefore, in the present study, we investigated the role of Siah2 in the IPC-induced neuroprotection in in vitro and in vivo models of IPC. To this aim, cortical neurons were exposed to 30-min oxygen and glucose deprivation (OGD, sublethal insult) followed by 3 h OGD plus reoxygenation (lethal insult). Our results revealed that the mitochondrial depolarization induced by hypoxia activates Siah2 at the mitochondrial level and increases LC3-II protein expression, a marker of mitophagy, an effect counteracted by the reoxygenation phase. By contrast, hypoxia reduced the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a marker of mitochondrial biogenesis, whereas its expression was increased after reoxygenation thus improving mitochondrial membrane potential, mitochondrial calcium content, and mitochondrial morphology, hence leading to neuroprotection in IPC. Furthermore, Siah2 silencing confirmed these results. Collectively, these findings indicate that the balance between mitophagy and mitochondrial biogenesis, due to the activation of the Siah2-E3-ligase, might play a role in IPC-induced neuroprotection.
Collapse
Affiliation(s)
- Maria Josè Sisalli
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Elena D'Apolito
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Giovanna Lombardi
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | - Michele Tufano
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy
| | | | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
38
|
Wang X, Ma C, Mi K, Cao X, Tan Y, Yuan H, Ren J, Liang X. Urolithin A attenuates Doxorubicin-induced cardiotoxicity by enhancing PINK1-regulated mitophagy via Ambra1. Chem Biol Interact 2025; 406:111363. [PMID: 39725191 DOI: 10.1016/j.cbi.2024.111363] [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: 09/23/2024] [Revised: 11/02/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Doxorubicin (Dox) is a widely used antineoplastics although its clinical usage is greatly limited by its cardiotoxicity. Several studies have depicted an essential role for dampened mitophagy and mitochondrial injury in Dox cardiotoxicity. However, preventative measure to alleviate Dox-evoked cardiotoxicity via targeting mitophagy and mitochondrial integrity remains elusive. Urolithin A (UA) is a newly identified mitophagy inducer with antioxidant and anti-apoptotic properties although its effect on Dox-induced cardiotoxicity is unknown. This study was designed to explore the effect of UA on Dox cardiotoxicity and mechanisms involved. Our results indicated that UA alleviated Dox-induced cardiac dysfunction exhibited by echocardiographic parameters and histological analyses, and partially relieved Dox-induced apoptosis in vitro and in vivo, and mitochondrial dysfunction including ΔΨm dissipation and ROS production in vitro. The ability of UA to facilitate restoration of mitophagy in mice and H9C2s underscored its advantageous effects, manifested as upregulation of mitophagy-related proteins, including p62, LC3, PINK1 and Parkin, as well as the co-location between LC3 and mitochondria. Incubation with 3 -MA nearly reversed the UA-evoked rise of mitophagy-related proteins, and inhibition of apoptosis. Given that knockdown of Ambra1 almost abolished UA-induced protective effect, the enhanced expression of Ambra1 owing to UA increased PINK1 levels by inhibiting its degradation via LONP1. Collectively, our results suggest that the cardioprotective properties of UA depend on the stimulation of PINK1-dependent mitophagy through promoting Ambra1 expression to inhibit PINK1 degradation by LONP1. This highlights UA's potential as a valuable treatment option and its importance in cardioprotective strategies against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Chao Ma
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Keying Mi
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Xinran Cao
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Yinghua Tan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Xinyue Liang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| |
Collapse
|
39
|
Ravi R, Routray D, Mahalakshmi R. Mitochondrial Sorting and Assembly Machinery: Chaperoning a Moonlighting Role? Biochemistry 2025; 64:312-328. [PMID: 39754567 DOI: 10.1021/acs.biochem.4c00727] [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] [Indexed: 01/06/2025]
Abstract
The mitochondrial outer membrane (OMM) β-barrel proteins link the mitochondrion with the cytosol, endoplasmic reticulum, and other cellular membranes, establishing cellular homeostasis. Their active insertion and assembly in the outer mitochondrial membrane is achieved in an energy-independent yet highly effective manner by the Sorting and Assembly Machinery (SAM) of the OMM. The core SAM constituent is the 16-stranded transmembrane β-barrel Sam50. For over two decades, the primary role of Sam50 has been linked to its function as a chaperone in the OMM, wherein it assembles all β-barrels through a lateral gating and β-barrel switching mechanism. Interestingly, recent studies have demonstrated that despite its low copy number, Sam50 performs various diverse functions beyond assembling β-barrels. This includes maintaining cristae morphology, bidirectional lipid shuttling between the ER and mitochondrial inner membrane, import of select proteins, regulation of PINK1-Parkin function, and timed trigger of cell death. Given these multifaceted critical regulatory functions of SAM across all eukaryotes, we now reason that SAM merely moonlights as the hub for β-barrel biogenesis and has indeed evolved a diverse array of primary roles in maintaining mitochondrial function and cellular homeostasis.
Collapse
Affiliation(s)
- Roshika Ravi
- Molecular Biophysics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| | - Deepsikha Routray
- Molecular Biophysics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| | - Radhakrishnan Mahalakshmi
- Molecular Biophysics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| |
Collapse
|
40
|
Shaikh K, Bowman M, McCormick SM, Gao L, Zhang J, White J, Tawil J, Kapoor A, Arav-Boger R, Norbury CC, Harhaj EW. ZFAND6 promotes TRAF2-dependent mitophagy to restrain cGAS-STING signaling. iScience 2025; 28:111544. [PMID: 39811672 PMCID: PMC11731517 DOI: 10.1016/j.isci.2024.111544] [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: 08/02/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
ZFAND6 is a zinc finger protein that interacts with TNF receptor-associated factor 2 (TRAF2) and polyubiquitin chains and has been linked to tumor necrosis factor (TNF) signaling. Here, we report a previously undescribed function of ZFAND6 in maintaining mitochondrial homeostasis by promoting mitophagy. Deletion of ZFAND6 in bone marrow-derived macrophages (BMDMs) upregulates reactive oxygen species (ROS) and the accumulation of damaged mitochondria due to impaired mitophagy. Consequently, mitochondrial DNA (mtDNA) is released into the cytoplasm, triggering the spontaneous expression of interferon-stimulated genes (ISGs) in a stimulator of interferon genes (STING) dependent manner, which leads to enhanced viral resistance. Mechanistically, ZFAND6 bridges a TRAF2-cIAP1 interaction and mediates the recruitment of TRAF2 to damaged mitochondria, which is required for the initiation of ubiquitin-dependent mitophagy. Our results suggest that ZFAND6 promotes the interactions of TRAF2 and cIAP1 and the clearance of damaged mitochondria by mitophagy to maintain mitochondrial homeostasis.
Collapse
Affiliation(s)
- Kashif Shaikh
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Melissa Bowman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Sarah M. McCormick
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Linlin Gao
- Department of Microbiology and Immunology, The University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Jiawen Zhang
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Jesse White
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - John Tawil
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Arun Kapoor
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Ravit Arav-Boger
- Department of Pediatrics, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Christopher C. Norbury
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Edward W. Harhaj
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
41
|
Xu W, Dong L, Dai J, Zhong L, Ouyang X, Li J, Feng G, Wang H, Liu X, Zhou L, Xia Q. The interconnective role of the UPS and autophagy in the quality control of cancer mitochondria. Cell Mol Life Sci 2025; 82:42. [PMID: 39800773 PMCID: PMC11725563 DOI: 10.1007/s00018-024-05556-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: 10/08/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Uncontrollable cancer cell growth is characterized by the maintenance of cellular homeostasis through the continuous accumulation of misfolded proteins and damaged organelles. This review delineates the roles of two complementary and synergistic degradation systems, the ubiquitin-proteasome system (UPS) and the autophagy-lysosome system, in the degradation of misfolded proteins and damaged organelles for intracellular recycling. We emphasize the interconnected decision-making processes of degradation systems in maintaining cellular homeostasis, such as the biophysical state of substrates, receptor oligomerization potentials (e.g., p62), and compartmentalization capacities (e.g., membrane structures). Mitochondria, the cellular hubs for respiration and metabolism, are implicated in tumorigenesis. In the subsequent sections, we thoroughly examine the mechanisms of mitochondrial quality control (MQC) in preserving mitochondrial homeostasis in human cells. Notably, we explored the relationships between mitochondrial dynamics (fusion and fission) and various MQC processes-including the UPS, mitochondrial proteases, and mitophagy-in the context of mitochondrial repair and degradation pathways. Finally, we assessed the potential of targeting MQC (including UPS, mitochondrial molecular chaperones, mitochondrial proteases, mitochondrial dynamics, mitophagy and mitochondrial biogenesis) as cancer therapeutic strategies. Understanding the mechanisms underlying mitochondrial homeostasis may offer novel insights for future cancer therapies.
Collapse
Affiliation(s)
- Wanting Xu
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Ji Dai
- Institute of International Technology and Economy, Development Research Center of the State Council, Beijing, 102208, China
| | - Lu Zhong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Xiao Ouyang
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Jiaqian Li
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Gaoqing Feng
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Huahua Wang
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Xuan Liu
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Liying Zhou
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
42
|
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
|
43
|
Tam E, Ouimet M, Sweeney G. Cardioprotective Effects of Adiponectin-Stimulated Autophagy. J Lipid Atheroscler 2025; 14:40-53. [PMID: 39911962 PMCID: PMC11791421 DOI: 10.12997/jla.2025.14.1.40] [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: 05/14/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 02/07/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure, pose a significant economic and health burden worldwide. Current treatment strategies for heart failure are greatly limited, in that they mainly mitigate symptoms or delay further progression. In contrast, therapies aimed at proactively preventing the onset of heart failure could greatly improve outcomes. Adiponectin is an adipocyte-derived hormone that confers an array of cardioprotective effects. It exerts anti-inflammatory effects, improves metabolic function, mitigates endothelial cell dysfunction, and reduce cardiomyocyte cell death. Furthermore, it has gained increasing attention for its ability to activate autophagy, a conserved cellular pathway that facilitates the degradation and recycling of cell components. The disruption of autophagy has been linked to CVDs including heart failure. Additionally, growing evidence also points to specific forms of autophagy, namely mitophagy and lipophagy, as crucial adaptive responses in protection against CVDs. The protective effects of adiponectin, autophagy, mitophagy, and lipophagy against CVDs along with potential therapeutic implications will be discussed.
Collapse
Affiliation(s)
- Eddie Tam
- Department of Biology, York University, Toronto, ON, Canada
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
44
|
Zheng C, Nguyen KK, Vishnivetskiy SA, Gurevich VV, Gurevich EV. Arrestin-3 binds parkin and enhances parkin-dependent mitophagy. J Neurochem 2025; 169:e16043. [PMID: 38196269 PMCID: PMC11231064 DOI: 10.1111/jnc.16043] [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: 05/08/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024]
Abstract
Arrestins were discovered for their role in homologous desensitization of G-protein-coupled receptors (GPCRs). Later non-visual arrestins were shown to regulate several signaling pathways. Some of these pathways require arrestin binding to GPCRs, the regulation of others is receptor independent. Here, we demonstrate that arrestin-3 binds the E3 ubiquitin ligase parkin via multiple sites, preferentially interacting with its RING0 domain. Identification of the parkin domains involved suggests that arrestin-3 likely relieves parkin autoinhibition and/or stabilizes the enzymatically active "open" conformation of parkin. Arrestin-3 binding enhances ubiquitination by parkin of the mitochondrial protein mitofusin-1 and facilitates parkin-mediated mitophagy in HeLa cells. Furthermore, arrestin-3 and its mutant with enhanced parkin binding rescue mitofusin-1 ubiquitination and mitophagy in the presence of the Parkinson's disease-associated R275W parkin mutant, which is defective in both functions. Thus, modulation of parkin activity via arrestin-3 might be a novel strategy of anti-parkinsonian therapy.
Collapse
Affiliation(s)
- Chen Zheng
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Kevin K. Nguyen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
45
|
Zhang JX, Lan MF, Shang JZ, Lai XL, Li LS, Duan TT, Xu RH, Chen KL, Duan X. DMT1 Maintains Iron Homeostasis to Regulate Mitochondrial Function in Porcine Oocytes. J Cell Physiol 2025; 240:e31494. [PMID: 39639679 DOI: 10.1002/jcp.31494] [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: 06/30/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Iron plays critical roles in many cellular functions, including energy production, metabolism, and cell proliferation. However, the role of iron in maintaining oocyte quality remains unclear. In this study, DMT1 was identified as a key iron transporter during porcine oocyte maturation. The results demonstrated that iron deficiency in porcine oocyte led to aberrant meiotic progression, accompanied by increased gene expression of DMT1. Inhibition of DMT1 resulted in the failure of cumulus cell expansion and oocyte maturation, along by the abnormal actin and microtubule assembly. Furthermore, loss of DMT1 function caused disruption in mitochondrial function and dynamics, resulting in oxidative stress and Ca2+ dyshomeostasis. Additionally, the absence of DMT1 function activated PINK1/Parkin-dependent mitophagy in porcine oocyte. These findings suggested that DMT1 played a crucial role in safeguarding oocyte quality by protecting against iron-deficiency-induced mitochondrial dysfunction and autophagy. This study provided compelling evidence that DMT1 and iron homeostasis were crucial for maintaining the capacity of porcine oocyte maturation. Moreover, the results hinted at the potential of DMT1 as a novel therapeutic target for treating iron deficiency-related female reproductive disorders.
Collapse
Affiliation(s)
- Jin-Xin Zhang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Meng-Fan Lan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Jian-Zhou Shang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Xin-Le Lai
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Li-Shu Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Tong-Tong Duan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
| | - Ru-Hai Xu
- Key Laboratory of Animal Genetics and Breeding of Zhejiang Province, Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Kun-Lin Chen
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, China
| | - Xing Duan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, Zhejiang, China
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
46
|
Zhou JX, Zheng ZY, Peng ZX, Yang YT, Ni HG. Predictive model in silicon and pathogenicity mechanism of metabolic syndrome: Impacts of heavy metal exposure. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 373:124001. [PMID: 39746257 DOI: 10.1016/j.jenvman.2024.124001] [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: 08/29/2024] [Revised: 12/03/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Although the association between heavy metals in human and the development of metabolic syndrome (MetS) have been extensively studied, the pathogenic mechanism of MetS affected by metals is not clear to date. In this study, a predictive model was developed with machine learning base on the large-scale dataset. These proposed models were evaluated via comparatively analysis of their accuracy and robustness. With the optimal model, two metals significantly correlated with MetS were screened and were employed to infer the pathogenicity mechanism of MetS via molecular docking. Significant associations between heavy metals and MetS were found. Molecular docking provided insights into the interactions between metal ions and key protein receptors involved in metabolic regulation, suggesting a mechanism by which heavy metals interfere with metabolic functions. Specifically, Ba and Cd affect the development of MetS thru their interactions with insulin and estrogen receptors. This study attempted to explore heavy metals' potential roles in MetS at the molecular level. These findings emphasize the importance of addressing environmental exposures in the prevention and treatment of MetS.
Collapse
Affiliation(s)
- Jing-Xuan Zhou
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zi-Yi Zheng
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zhao-Xing Peng
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Yu-Ting Yang
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Hong-Gang Ni
- School of Urban Planning and Design, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
47
|
Xu S, Wang Z, Guo F, Zhang Y, Peng H, Zhang H, Liu Z, Cao C, Xin G, Chen YY, Fu J. Mitophagy in ischemic heart disease: molecular mechanisms and clinical management. Cell Death Dis 2024; 15:934. [PMID: 39737905 DOI: 10.1038/s41419-024-07303-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025]
Abstract
The influence of the mitochondrial control system on ischemic heart disease has become a major focus of current research. Mitophagy, as a very crucial part of the mitochondrial control system, plays a special role in ischemic heart disease, unlike mitochondrial dynamics. The published reviews have not explored in detail the unique function of mitophagy in ischemic heart disease, therefore, the aim of this paper is to summarize how mitophagy regulates the progression of ischemic heart disease. We conclude that mitophagy affects ischemic heart disease by promoting cardiomyocyte hypertrophy and fibrosis, the progression of oxidative stress, the development of inflammation, and cardiomyocyte death, and that the specific mechanisms of mitophagy are worthy of further investigation.
Collapse
Affiliation(s)
- Shujuan Xu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110122, China
| | - Fan Guo
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yehao Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Han Peng
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Huiyu Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zixin Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Ce Cao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Gaojie Xin
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yuan Yuan Chen
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jianhua Fu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
48
|
Wu W, Wei Z, Wu Z, Chen J, Liu J, Chen M, Yuan J, Zheng Z, Zhao Z, Lin Q, Liu N, Chen H. Exercise training alleviates neuronal apoptosis and re-establishes mitochondrial quality control after cerebral ischemia by increasing SIRT3 expression. Cell Biol Toxicol 2024; 41:10. [PMID: 39707047 PMCID: PMC11662049 DOI: 10.1007/s10565-024-09957-3] [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: 06/26/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Existing evidence indicates that exercise training can enhance neural function by regulating mitochondrial quality control (MQC), which can be impaired by cerebral ischemia, and that sirtuin-3 (SIRT3), a protein localized in mitochondria, is crucial in maintaining mitochondrial functions. However, the relationship among exercise training, SIRT3, and MQC after cerebral ischemia remains obscure. This study attempted to elucidate the relationship among exercise training, SIRT3 and MQC after cerebral ischemia in rats. Male adult SD rats received tMCAO after the transfection of adeno-associated virus encoding either sirtuin-3 (AAV-SIRT3) or SIRT3 knockdown (AAV-sh-SIRT3) into the ipsilateral striata and cortex. Subsequently, the animals were randomly selected for exercise training. The index changes were measured by transmission electron microscopy, Western blot analysis, nuclear magnetic resonance imaging, TUNEL staining, and immunofluorescence staining, etc. The results revealed that after cerebral ischemia, exercise training increased SIRT3 expression, significantly improved neural function, alleviated infarct volume and neuronal apoptosis, maintained the mitochondrial structural integrity, and re-established MQC. The latter promoted mitochondrial biogenesis, balanced mitochondrial fission/fusion, and enhanced mitophagy. These favorable benefits were reversed after SIRT3 interference. In addition, a cellular OGD/R model showed that the increased SIRT3 expression alleviates neuronal apoptosis and re-establishes mitochondrial quality control by activating the β-catenin pathway. These findings suggest that exercise training may optimize mitochondrial quality control by increasing the expression of SIRT3, thereby improving neural functions after cerebral ischemia, which illuminates the mechanism underlying the exercise training-conferred neural benefits and indicates SIRT3 as a therapeutic strategy for brain ischemia.
Collapse
Affiliation(s)
- Wenwen Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zengyu Wei
- Emergency Department, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhiyun Wu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Jianmin Chen
- Department of Rehabilitation Medicine, The First Afliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Jinjin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zhijian Zheng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Zijun Zhao
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Qiang Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China.
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
49
|
Chu H, Zhu H, Ma J, Jiang Y, Cui C, Yan X, Li Q, Zhang X, Chen D, Li X, Li R. Mitochondrial Dysfunction and Metabolic Indicators in Patients with Drug-Naive First-Episode Schizophrenia: A Case-Control Study. Neuropsychiatr Dis Treat 2024; 20:2433-2442. [PMID: 39687781 PMCID: PMC11647910 DOI: 10.2147/ndt.s501527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024] Open
Abstract
Objective This paper aims to explore the expression characteristics of mitochondrial function-related genes in patients with first-episode schizophrenia (SCZ)and the correlation between differentially expressed genes and clinical metabolic indicators. Methods Twenty patients with first-episode SCZ who had not taken antipsychotic drugs (patient group) and twenty healthy controls (control group) were included. Quantitative real-time PCR technology was used to detect the expression levels of genes related to mitochondrial quality control and oxidative phosphorylation in peripheral blood leukocytes, and metabolic indicators such as blood biochemistry and blood glucose were collected. Results The gene expression levels of key genes related to mitochondrial function, PGC-1a, PARK2, and LC3B, in the patient group were significantly lower than those in the control group (P < 0.05). Correlation analysis showed that the expression level of PGC-1a gene in the patient group was negatively correlated with very low-density lipoprotein levels (r =-0.451), and the expression level of PARK2 gene in the patient group was negatively correlated with uric acid levels (r =-0.447). Conclusion The expression levels of multiple key genes in the mitochondrial quality control and oxidative phosphorylation processes in patients with first-episode SCZ display a downward trend. The differentially expressed genes are correlated with the metabolic abnormalities of the patients, suggesting that mitochondrial dysfunction may be related to the high incidence of metabolic diseases in patients with SCZ.
Collapse
Affiliation(s)
- Haoran Chu
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Houming Zhu
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Jiashu Ma
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Yijia Jiang
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Cuicui Cui
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Xianxia Yan
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Qin Li
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Xinyan Zhang
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Diancai Chen
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Xianwei Li
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| | - Ranran Li
- Department of Psychiatry, Shandong Mental Health Center, Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
50
|
Chaphalkar RM, Kodati B, Maddineni P, He S, Brooks CD, Stankowska DL, Yang S, Zode G, Krishnamoorthy RR. A Reduction in Mitophagy Is Associated with Glaucomatous Neurodegeneration in Rodent Models of Glaucoma. Int J Mol Sci 2024; 25:13040. [PMID: 39684751 PMCID: PMC11642561 DOI: 10.3390/ijms252313040] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Glaucoma is a heterogenous group of optic neuropathies characterized by the degeneration of optic nerve axons and the progressive loss of retinal ganglion cells (RGCs), which could ultimately lead to vision loss. Elevated intraocular pressure (IOP) is a major risk factor in the development of glaucoma, and reducing IOP remains the main therapeutic strategy. Endothelin-1 (ET-1), a potent vasoactive peptide, has been shown to produce neurodegenerative effects in animal models of glaucoma. However, the detailed mechanisms underlying ET-1-mediated neurodegeneration in glaucoma are not completely understood. In the current study, using a Seahorse Mitostress assay, we report that ET-1 treatment for 4 h and 24 h time points causes a significant decline in various parameters of mitochondrial function, including ATP production, maximal respiration, and spare respiratory capacity in cultured RGCs. This compromise in mitochondrial function could trigger activation of mitophagy as a quality control mechanism to restore RGC health. Contrary to our expectation, we observed a decrease in mitophagy following ET-1 treatment for 24 h in cultured RGCs. Using Morrison's model of ocular hypertension in rats, we investigated here, for the first time, changes in mitophagosome formation by analyzing the co-localization of LC-3B and TOM20 in RGCs. We also injected ET-1 (24 h) into transgenic GFP-LC3 mice to analyze the formation of mitophagosomes in vivo. In Morrison's model of ocular hypertension, as well as in ET-1 injected GFP-LC3 mice, we found a decrease in co-localization of LC3 and TOM20, indicating reduced mitophagy. Taken together, these results demonstrate that both ocular hypertension and ET-1 administration in rats and mice lead to reduced mitophagy, thus predisposing RGCs to neurodegeneration.
Collapse
Affiliation(s)
- Renuka M. Chaphalkar
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.M.C.); (B.K.); (C.D.B.); (S.Y.)
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Bindu Kodati
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.M.C.); (B.K.); (C.D.B.); (S.Y.)
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Prabhavathi Maddineni
- Department of Ophthalmology, School of Medicine, University of Missouri, Columbia, MO 65212, USA;
| | - Shaoqing He
- Department of Pathology, Children’s Health at Dallas, Dallas, TX 75235, USA;
| | - Calvin D. Brooks
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.M.C.); (B.K.); (C.D.B.); (S.Y.)
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Dorota L. Stankowska
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
- Department of Microbiology, Immunology and Genetics, College of Biomedical and Translational Sciences at University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shaohua Yang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.M.C.); (B.K.); (C.D.B.); (S.Y.)
| | - Gulab Zode
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California, Irvine, CA 92697, USA;
| | - Raghu R. Krishnamoorthy
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (R.M.C.); (B.K.); (C.D.B.); (S.Y.)
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| |
Collapse
|