1
|
Yerra VG, Connelly KA. MOTS-c: Magical Molecule for Diabetic Cardiomyopathy? Cardiovasc Drugs Ther 2025; 39:473-476. [PMID: 40172798 DOI: 10.1007/s10557-025-07689-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Affiliation(s)
- Veera Ganesh Yerra
- Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 bond St, Toronto, ON, M5B1W8, Canada
| | - Kim A Connelly
- Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, 30 bond St, Toronto, ON, M5B1W8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Iboleon-Jimenez A, Sánchez-Quintero MJ, Carmona-Segovia ADM, Sojo B, Fernández-Ramos AM, García-Rodríguez L, Molina-Ramos AI, García-Pinilla JM, Jimenez-Navarro M, Ortega-Gomez A. Circulating mitochondrial biomarkers in acute coronary syndrome. Front Med (Lausanne) 2025; 12:1568305. [PMID: 40443515 PMCID: PMC12119305 DOI: 10.3389/fmed.2025.1568305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/29/2025] [Indexed: 06/02/2025] Open
Abstract
Background Acute coronary syndrome (ACS) is the leading cause of mortality in developed countries. Mitochondrial dysfunction is a hallmark of various cardiometabolic diseases, including ACS. Emerging evidence suggests that evaluating mitochondrial biomarkers in plasma may offer valuable insights into the pathophysiology and management of these conditions. The present study aims to analyse the effect of ACS, sex and their interaction on plasma levels of mitochondrial markers, such as peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), mitochondrial open reading frame of the 12S rRNA type-c (MOTS-c) and citrate syntetase (CS). Methods A total of 18 ACS patients (8 women and 10 men) and 20 controls (8 women and 12 men) were included in this study. Venous blood samples were collected from participants after a 12-h overnight fast. Plasma levels of mitochondrial PGC-1α, MOTS-c and CS were measured. Results ACS significantly reduced plasma levels of PGC-1α and MOTS-c. Sex did not shown a significant effect on these markers. Additionally, MOTS-c positively correlated with the first troponin and hemoglobin, PGC-1α negatively correlated with glucose and positively with HDL-cholesterol, and CS showed negative correlations with NT-proBNP, C-reactive protein, and hemoglobin. Conclusion Mitochondria markers, MOTS-c and PGC-1α, are altered in ACS patients, with no observed sex differences. These findings represent an initial step toward integrating personalized medicine into the clinical management of ACS. Nonetheless, further studies are required to fully elucidate the role of these markers in this pathology.
Collapse
Affiliation(s)
- Andrea Iboleon-Jimenez
- Unidad Docente Multiprofesional de Atención Familiar y Comunitaria, Distrito de Atención Primaria Málaga-Guadalhorce; Faculty of Medicine, University of Málaga, Málaga, Spain
| | - María J. Sánchez-Quintero
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Ada D. M. Carmona-Segovia
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Bélen Sojo
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Endocrinology and Nutrition UGC, Victoria Virgen University Hospital, Málaga, Spain
| | - Ana María Fernández-Ramos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Clinical Analysis UGC, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Luis García-Rodríguez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
| | - Ana I. Molina-Ramos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - José Manuel García-Pinilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Málaga, Spain
| | - Manuel Jimenez-Navarro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Department of Cardiology and Cardiovascular Surgery, Virgen de la Victoria University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Málaga, Spain
| | - Almudena Ortega-Gomez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga, Spain
- Endocrinology and Nutrition UGC, Victoria Virgen University Hospital, Málaga, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Ajala I, Vanderperre B. Non-canonical ORFs-derived protein products in mitochondria: A multifaceted exploration of their functions in health and disease. Protein Sci 2025; 34:e70053. [PMID: 39969119 PMCID: PMC11837024 DOI: 10.1002/pro.70053] [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: 10/09/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/20/2025]
Abstract
Traditionally, eukaryotic mRNAs were perceived as inherently monocistronic. However, recent insights from ribosome profiling (Ribo-seq) and proteomics studies challenge this paradigm. These investigations reveal that, beyond the currently annotated reference proteins (RefProts), there exist additional proteins known as alternative proteins (AltProts) and small open reading frames derived microproteins encoded in regions of mRNAs previously considered untranslated or in non-coding transcripts. This experimental evidence broadens the spectrum of functional proteins within cells, tissues, and organs, potentially offering crucial insights into biological processes. Notably, a significant proportion of these newly identified AltProts and microproteins demonstrates localization in mitochondria, contributing to the functions of mitochondrial complexes. This review delves into the overlooked realm of the alternative proteome within mitochondria, exploring the role of nuclear or mitochondrial-genome-encoded AltProts and microproteins in physiological and pathological cellular processes.
Collapse
Affiliation(s)
- Ikram Ajala
- Department of Biological Sciences, Université du Québec à MontréalCERMO‐FC Research CenterMontrealQuebecCanada
- Network for Research on Protein FunctionEngineering and Applications (PROTEO)MontréalQuebecCanada
| | - Benoît Vanderperre
- Department of Biological Sciences, Université du Québec à MontréalCERMO‐FC Research CenterMontrealQuebecCanada
- Network for Research on Protein FunctionEngineering and Applications (PROTEO)MontréalQuebecCanada
| |
Collapse
|
4
|
Mohtashami Z, Schneider K, Azimi R, Atilano S, Chwa M, Kenney MC, Singh MK. Exploring the therapeutic potential of MOTS-c in age-related macular degeneration: from cellular responses to patient-derived cybrids. Hum Cell 2025; 38:57. [PMID: 39961901 DOI: 10.1007/s13577-025-01188-w] [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: 09/08/2024] [Accepted: 02/11/2025] [Indexed: 05/09/2025]
Abstract
Age-related macular degeneration (AMD), the leading cause of irreversible vision loss in the US, is on the rise among the elderly. Uncontrolled mitochondria-derived peptide production from mtDNA disruption and 16S or 12S rRNA damage could worsen AMD. Our previous work has shown that Humanin G possesses cytoprotective effects in retinal pigment epithelial (RPE) cells. However, MOTS-c, a highly efficient mitochondrial peptide, has yet to be evaluated on retinal cell survival. In this study, we show that there are differences in effects between wild-type (wt-) and differentiated ARPE19 cells (diff-ARPE19), implying that the cellular differentiation status may influence how cells respond to MOTS-c. MOTS-c has dose-dependent effects on apoptosis, inflammation, and mitochondrial biogenesis in diff-ARPE19 cells. Lower doses (500 nM) have more significant impacts than 5 µM concentrations. In diff-ARPE19 cells, a lower dose of MOTS-c can reduce the negative impact of hypoxia on cellular survival and gene expression, including apoptosis (CASP3, CASP9), mitochondrial biogenesis (TFAM, PGC-1α), and metabolic sensor (AMPK). However, it had no significant effect on ROS levels or NRF1 expression, regardless of MOTS-c dose. Exposing diff-ARPE19 cells to varied MOTS-c dosages before and after therapy in a chemically induced hypoxic environment yields no extra benefits as compared to MOTS-c treatment alone. MOTS-c had different effects on the expression of genes linked with apoptosis, mitochondrial biogenesis, and antioxidant activity in AMD patients versus age-matched control cybrids. The MOTS-c peptide appears to enhance cellular metabolism and regulate gene expression, which could potentially provide therapeutic benefits in AMD.
Collapse
Affiliation(s)
- Zahra Mohtashami
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA.
| | - Kevin Schneider
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - Reza Azimi
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - Shari Atilano
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - Marilyn Chwa
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
| | - M Cristina Kenney
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Mithalesh Kumar Singh
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, 92697, USA.
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA.
| |
Collapse
|
5
|
Zicarelli M, Greco M, Roumeliotis S, Lo Vasco ME, Dragone F, Kourtidou C, Alekos I, Misiti R, Foti DP, Coppolino G, Liakopoulos V, Dounousi E, Bolignano D. MOTS-c Levels and Sarcopenia Risk in Chronic Peritoneal Dialysis Patients: A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:322. [PMID: 40005438 PMCID: PMC11857303 DOI: 10.3390/medicina61020322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Sarcopenia is exceedingly frequent in end-stage kidney disease (ESKD) patients on dialysis, including those undergoing peritoneal dialysis (PD), and is of multifactorial origin. MOTS-c is a mitochondrial-derived peptide that promotes muscle growth whose levels are unbalanced in ESKD. In this study, we evaluated MOTS-c balance and its relationship with sarcopenia risk in an ESKD-PD cohort. Materials and Methods: MOTS-c was measured in serum, urine, and dialysate samples of 32 chronic PD patients. Patients were thus screened for sarcopenia risk by the SARC-F tool, anthropometric measurements, and physical performance tests. Results: PD patients with a very high sarcopenia risk (SARC-F ≥ 2) had significantly lower serum (sMOTS-c) and higher dialysate (dMOTS-c) levels, suggesting an increased peritoneal clearance of this substance (d/s MOTS-c). sMOTS-c levels were directly correlated with muscle performance in physical tests, while an opposite relationship was found with dMOTS-c and d/sMOTS-c. ROC analyses demonstrated the diagnostic potential of MOTS-c, particularly in combination with physical and anthropometric assessments, to identify PD patients at very high risk of sarcopenia. Conclusions: Chronic PD may negatively affect MOTS-c balance, which, in turn, may contribute to enhanced sarcopenia risk. Larger studies are needed to confirm these observations and to validate the potential utility of this substance as a biomarker for improving sarcopenia risk stratification in PD patients.
Collapse
Affiliation(s)
| | - Marta Greco
- Department of Health Sciences, Magna-Graecia University, 88100 Catanzaro, Italy
- Clinical Pathology Lab, Magna-Graecia University Hospital, 88100 Catanzaro, Italy
| | - Stefanos Roumeliotis
- 2nd Department of Nephrology, AHEPA University Hospital Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | | | - Francesco Dragone
- Clinical Pathology Lab, Magna-Graecia University Hospital, 88100 Catanzaro, Italy
| | - Christodoula Kourtidou
- 2nd Department of Nephrology, AHEPA University Hospital Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Ioannis Alekos
- Department of Nephrology, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Roberta Misiti
- Clinical Pathology Lab, Magna-Graecia University Hospital, 88100 Catanzaro, Italy
| | - Daniela Patrizia Foti
- Clinical Pathology Lab, Magna-Graecia University Hospital, 88100 Catanzaro, Italy
- Department of Experimental and Clinical Medicine, Magna-Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Coppolino
- Department of Health Sciences, Magna-Graecia University, 88100 Catanzaro, Italy
| | - Vassilios Liakopoulos
- 2nd Department of Nephrology, AHEPA University Hospital Medical School, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Evangelia Dounousi
- Department of Nephrology, School of Medicine, University of Ioannina, 451 10 Ioannina, Greece
| | - Davide Bolignano
- Department of Medical and Surgical Sciences, Magna-Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
6
|
Feng Y, Rao Z, Tian X, Hu Y, Yue L, Meng Y, Zhong Q, Chen W, Xu W, Li H, Hu Y, Shi R. Endurance training enhances skeletal muscle mitochondrial respiration by promoting MOTS-c secretion. Free Radic Biol Med 2025; 227:619-628. [PMID: 39706498 DOI: 10.1016/j.freeradbiomed.2024.12.038] [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: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
The mitochondrial open reading frame of 12S rRNA-c (MOTS-c) is a biologically active mitochondria-derived peptide. However, the relationship between MOTS-c, skeletal muscle mitochondrial function, and endurance exercise adaptations is unknown. Here, we tested indices such as maximal oxygen uptake and serum MOTS-c levels in marathon runners and sedentary subjects. In addition, we tested aerobic exercise capacity, skeletal muscle mitochondrial respiration rate, and serum MOTS-c levels in mice subjected to long-term endurance training groups and sedentary groups. Our results indicated a close association between serum MOTS-c levels and aerobic exercise capacity. Circulating MOTS-c levels are expected to be an important indicator for predicting aerobic exercise capacity and assessing body fat status, endurance training load, and physical function. More importantly, we found that endurance training may enhance the mitochondrial respiratory function of skeletal muscle by promoting the secretion of MOTS-c and activating the AMPK/PGC-1α pathway.
Collapse
Affiliation(s)
- Yiwei Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Zhijian Rao
- School of Physical Education, Shanghai Normal University, Shanghai, 200234, China
| | - Xu Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yi Hu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Liantian Yue
- School of Sport, Exercise and Health Sciences, Loughborough University, UK
| | - Yifan Meng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Qiuling Zhong
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Wei Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Wenlong Xu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Haoran Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yingjia Hu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Rengfei Shi
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
7
|
Shen H, Nie J, Wang X, Li G, Zhao L, Jin Y, Jin L. MOTS-c relieves hepatocellular carcinoma resistance to TRAIL-induced apoptosis under hypoxic conditions by activating MEF2A. Exp Cell Res 2025; 444:114354. [PMID: 39581216 DOI: 10.1016/j.yexcr.2024.114354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Mitochondrial ORF of the 12S rRNA type-c (MOTS-c) as an AMPK agonist can regulate the expression of adaptive nuclear genes to promote cell homeostasis. However, the investigation of MOTS-c in hepatocellular carcinoma (HCC) is insufficient. This study aims to reveal the role of MOTS-c on HCC cell apoptosis. METHODS Huh7 and HCCLM3 cells were incubated with MOTS-c under a hypoxic condition. MOTS-c levels were quantified by enzyme-linked immunosorbent assay in the peripheral blood of HCC patients and healthy controls. Cell viability was detected by 3-(4,5-Dimethylthazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell apoptosis was investigated by flow cytometry and Tunel assay. Protein expression was detected by western blotting or immunohistochemistry assay. Dual-luciferase reporter assay and chromatin immunoprecipitation assay were performed to identify the association among myocyte enhancer factor 2A (MEF2A), death receptor 4 (DR4) and DR5. A tumor-bearing nude mouse model was conducted to assess the effect of MOTS-c on HCC tumor formation in vivo. RESULTS MOTS-c levels in the peripheral blood of HCC patients were significantly lower compared to healthy individuals. MOTS-c promoted HCC cell apoptosis under hypoxia conditions. Hypoxia stimulation decreased the protein expression of MEF2A, DR4, DR5, fas-associating via death domain (FADD) and caspase-8, while these effects were attenuated after MOTS-c treatment. MOTS-c induced TRAIL-induced apoptosis of HCC cells by activating MEF2A through the phosphorylation of AMPK under hypoxia treatment. In addition, MEF2A transcriptionally up-regulated DR4 and DR5. MOTS-c activated MEF2A to regulate DR4 and DR5 expression, further mediating TRAIL-induced apoptosis. Further, MOTS-c treatment relieved hypoxia-induced tumor growth in vivo. CONCLUSION MOTS-c relieved hypoxia-induced HCC cell resistance to TRAIL-caused apoptosis by activating MEF2A.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/drug therapy
- Apoptosis/drug effects
- Animals
- Mice
- TNF-Related Apoptosis-Inducing Ligand/metabolism
- TNF-Related Apoptosis-Inducing Ligand/pharmacology
- MEF2 Transcription Factors/metabolism
- MEF2 Transcription Factors/genetics
- Mice, Nude
- Cell Line, Tumor
- Male
- Female
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Mice, Inbred BALB C
- Xenograft Model Antitumor Assays
- Middle Aged
- Cell Hypoxia/drug effects
Collapse
Affiliation(s)
- Haiying Shen
- Department of Pathophysiology, School of Basic Medicine, Jilin Medical University, Jilin 132013, Jilin Province, PR China.
| | - Junjie Nie
- Department of Nuclear Medicine, Jilin People's Hospital, Jilin 132001, Jilin Province, PR China
| | - Xiaojun Wang
- School of Public Health, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Guangqing Li
- Department of Computer Application, School of Biomedical Engineering, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Liwei Zhao
- Department of Pathology, School of Basic Medicine, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Yuji Jin
- Department of Medical Genetics, School of Basic Medicine, Jilin Medical University, Jilin 132013, Jilin Province, PR China
| | - Lianhai Jin
- Hypoxia and Health Medicine Research Center, Jilin Medical University, Jilin 132013, Jilin Province, PR China.
| |
Collapse
|
8
|
Kim S. The Relationship Between MOTS-c K14Q Polymorphism and Sarcopenia, Blood Lipids, and Mental Health in Older Korean Adults. Biomedicines 2024; 12:2384. [PMID: 39457696 PMCID: PMC11504729 DOI: 10.3390/biomedicines12102384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/objectives: An East Asian-specific 1382A>C polymorphism in the mitochondrial open reading frame of the 12S rRNA type-c results in an amino acid substitution from Lys (K) to Gln (Q) at the 14th amino acid residue. This study investigated the association between m.1382A>C polymorphism and sarcopenia, blood lipids, and mental health in older Korean adults. Methods: The study included 683 community-dwelling Korean adults (345 men and 338 women) aged 65 years and older. The m.1382A>C polymorphism was genotyped with a 7500 real-time PCR system. Handgrip strength (HGS) was measured, and appendicular skeletal muscle (ASM) mass was calculated. Demographics, blood lipids, falling risk, nutritional intake, cognition function, and depression were additionally measured. Results: Men carrying the C allele had significantly higher ASM (21.6 ± 3.0 vs. 19.5 ± 2.2 kg, p = 0.018), ASM/height2 (7.76 ± 0.76 vs. 7.14 ± 0.62 kg/m2, p = 0.012), lean mass (53.3 ± 6.2 vs. 46.5 ± 4.0 kg, p < 0.001), left HGS (33.3 ± 5.0 vs. 28.9 ± 4.0 kg, p = 0.010), and right HGS (35.6 ± 5.3 vs. 30.9 ± 4.3 kg, p = 0.009) than men carrying the A allele. The genotype differences in ASM (p = 0.017), ASM/height2 (p = 0.011), lean mass (p < 0.001), left HGS (p = 0.010), and right HGS (p = 0.009) remained significant even after adjusting for all measured covariates. By contrast, no significant differences in other measured parameters were found between women carrying the A and C alleles. Conclusions: Our study findings indicate that the m.1382A>C polymorphism may be used as a genetic biomarker of age-related sarcopenia in older Korean men.
Collapse
Affiliation(s)
- Shinuk Kim
- Gyedang College of General Education, Sangmyung University, Cheonan 31066, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
9
|
Nichols C, Do-Thi VA, Peltier DC. Noncanonical microprotein regulation of immunity. Mol Ther 2024; 32:2905-2929. [PMID: 38734902 PMCID: PMC11403233 DOI: 10.1016/j.ymthe.2024.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024] Open
Abstract
The immune system is highly regulated but, when dysregulated, suboptimal protective or overly robust immune responses can lead to immune-mediated disorders. The genetic and molecular mechanisms of immune regulation are incompletely understood, impeding the development of more precise diagnostics and therapeutics for immune-mediated disorders. Recently, thousands of previously unrecognized noncanonical microprotein genes encoded by small open reading frames have been identified. Many of these microproteins perform critical functions, often in a cell- and context-specific manner. Several microproteins are now known to regulate immunity; however, the vast majority are uncharacterized. Therefore, illuminating what is often referred to as the "dark proteome," may present opportunities to tune immune responses more precisely. Here, we review noncanonical microprotein biology, highlight recently discovered examples regulating immunity, and discuss the potential and challenges of modulating dysregulated immune responses by targeting microproteins.
Collapse
Affiliation(s)
- Cydney Nichols
- Morris Green Scholars Program, Department of Pediatrics, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Van Anh Do-Thi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Daniel C Peltier
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
10
|
Petrikonis K, Bernatoniene J, Kopustinskiene DM, Casale R, Davinelli S, Saso L. The Antinociceptive Role of Nrf2 in Neuropathic Pain: From Mechanisms to Clinical Perspectives. Pharmaceutics 2024; 16:1068. [PMID: 39204413 PMCID: PMC11358986 DOI: 10.3390/pharmaceutics16081068] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Neuropathic pain, a chronic condition resulting from nerve injury or dysfunction, presents significant therapeutic challenges and is closely associated with oxidative stress and inflammation, both of which can lead to mitochondrial dysfunction. The nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, a critical cellular defense mechanism against oxidative stress, has emerged as a promising target for neuropathic pain management. Nrf2 modulators enhance the expression of antioxidant and cytoprotective genes, thereby reducing oxidative damage, inflammation, and mitochondrial impairment. This review explores the antinociceptive effects of Nrf2, highlighting how pharmacological agents and natural compounds may be used as potential therapeutic strategies against neuropathic pain. Although preclinical studies demonstrate significant pain reduction and improved nerve function through Nrf2 activation, several clinical challenges need to be addressed. However, emerging clinical evidence suggests potential benefits of Nrf2 modulators in several conditions, such as diabetic neuropathy and multiple sclerosis. Future research should focus on further elucidating the molecular role of Nrf2 in neuropathic pain to optimize its modulation efficacy and maximize clinical utility.
Collapse
Affiliation(s)
- Kestutis Petrikonis
- Department of Neurology, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50009 Kaunas, Lithuania;
| | - Jurga Bernatoniene
- Department of Drug Technology and Social Pharmacy, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania;
| | - Dalia M. Kopustinskiene
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Medical Academy, Lithuanian University of Health Sciences, Sukileliu pr. 13, LT-50161 Kaunas, Lithuania;
| | - Roberto Casale
- Opusmedica Persons, Care & Research-NPO, 29121 Piacenza, Italy;
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, La Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
11
|
Bolignano D, Greco M, Presta P, Duni A, Zicarelli M, Mercuri S, Pappas E, Lakkas L, Musolino M, Naka KK, Misiti R, Foti DP, Andreucci M, Coppolino G, Dounousi E. The Mitochondrial-Derived Peptide MOTS-c May Refine Mortality and Cardiovascular Risk Prediction in Chronic Hemodialysis Patients: A Multicenter Cohort Study. Blood Purif 2024; 53:824-837. [PMID: 39111290 DOI: 10.1159/000540303] [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: 04/11/2024] [Accepted: 07/07/2024] [Indexed: 09/03/2024]
Abstract
INTRODUCTION Uremic patients exhibit remarkably increased rates of mortality and cardiovascular (CV) events, but risk prediction in this setting remains difficult. Systemic mitochondrial dysfunction is pervasive in end-stage kidney disease and may contribute to CV complications. We tested the clinical significance of circulating MOTS-c, a small mitochondrial-derived peptide, as a biomarker for improving mortality and CV risk prediction in hemodialysis (HD) patients. METHODS We conducted a prospective, observational, multicenter study on 94 prevalent HD patients. The study endpoint was a composite of all-cause mortality and non-fatal CV events. The diagnostic and prognostic capacities of predictive models based on cohort-related risk factors were tested before and after the inclusion of MOTS-c. RESULTS MOTS-c levels were higher in HD patients than in controls (p < 0.001) and even more elevated (p = 0.01) in the 53 individuals experiencing the combined endpoint during follow-up (median duration: 26.5 months). MOTS-c was independently associated with the endpoint at either multivariate logistic (OR 1.020; 95% CI: 1.011-1.109; p = 0.03) or Cox regression analyses (HR 1.004; 95% CI: 1.000-1.025; p = 0.05) and the addition of this biomarker to prognostic models including the other cohort-related risk predictors (age, left ventricular mass, evidence of diastolic dysfunction, diabetes, pulse pressure) significantly improved the calibration, risk variability explanation, discrimination (receiver operating characteristic area under the curve from 0.727 to 0.743; C-index from 0.658 to 0.700), and particularly, the overall reclassification capacity (NRI 15.87%; p = 0.01). CONCLUSIONS In HD patients, the mitochondrial-derived peptide MOTS-c may impart significant information to refine CV risk prediction, beyond cohort-related risk factors. Future investigations are needed to generalize these findings in larger and more heterogeneous cohorts.
Collapse
Affiliation(s)
- Davide Bolignano
- Nephrology and Dialysis Unit, Magna Graecia University, Catanzaro, Italy
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marta Greco
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
- Clinical Pathology Lab, Magna Graecia University, Catanzaro, Italy
| | - Pierangela Presta
- Nephrology and Dialysis Unit, Magna Graecia University, Catanzaro, Italy
| | - Anila Duni
- Department of Nephrology, School of Medicine, University of Ioannina, Ioannina, Greece
| | | | - Simone Mercuri
- Nephrology and Dialysis Unit, Magna Graecia University, Catanzaro, Italy
| | - Efthymios Pappas
- Hemodialysis Unit, General Hospital of Filiates, Filiates, Greece
| | - Lampros Lakkas
- Second Department of Cardiology, University Hospital of Ioannina, Ioannina, Greece
| | - Michela Musolino
- Nephrology and Dialysis Unit, Magna Graecia University, Catanzaro, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Katerina K Naka
- Second Department of Cardiology, University Hospital of Ioannina, Ioannina, Greece
| | - Roberta Misiti
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
- Clinical Pathology Lab, Magna Graecia University, Catanzaro, Italy
| | - Daniela Patrizia Foti
- Clinical Pathology Lab, Magna Graecia University, Catanzaro, Italy
- Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Michele Andreucci
- Nephrology and Dialysis Unit, Magna Graecia University, Catanzaro, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Giuseppe Coppolino
- Nephrology and Dialysis Unit, Magna Graecia University, Catanzaro, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Evangelia Dounousi
- Department of Nephrology, School of Medicine, University of Ioannina, Ioannina, Greece
| |
Collapse
|
12
|
Da W, Chen Q, Shen B. The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration. Biol Res 2024; 57:37. [PMID: 38824571 PMCID: PMC11143644 DOI: 10.1186/s40659-024-00494-1] [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: 07/31/2023] [Accepted: 04/03/2024] [Indexed: 06/03/2024] Open
Abstract
It is widely acknowledged that aging, mitochondrial dysfunction, and cellular phenotypic abnormalities are intricately associated with the degeneration of bone and cartilage. Consequently, gaining a comprehensive understanding of the regulatory patterns governing mitochondrial function and its underlying mechanisms holds promise for mitigating the progression of osteoarthritis, intervertebral disc degeneration, and osteoporosis. Mitochondrial hormesis, referred to as mitohormesis, represents a cellular adaptive stress response mechanism wherein mitochondria restore homeostasis and augment resistance capabilities against stimuli by generating reactive oxygen species (ROS), orchestrating unfolded protein reactions (UPRmt), inducing mitochondrial-derived peptides (MDP), instigating mitochondrial dynamic changes, and activating mitophagy, all prompted by low doses of stressors. The varying nature, intensity, and duration of stimulus sources elicit divergent degrees of mitochondrial stress responses, subsequently activating one or more signaling pathways to initiate mitohormesis. This review focuses specifically on the effector molecules and regulatory networks associated with mitohormesis, while also scrutinizing extant mechanisms of mitochondrial dysfunction contributing to bone and cartilage degeneration through oxidative stress damage. Additionally, it underscores the potential of mechanical stimulation, intermittent dietary restrictions, hypoxic preconditioning, and low-dose toxic compounds to trigger mitohormesis, thereby alleviating bone and cartilage degeneration.
Collapse
Affiliation(s)
- Wacili Da
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Quan Chen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bin Shen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
13
|
Yang L, Li M, Liu Y, Bai Y, Yin T, Chen Y, Jiang J, Liu S. MOTS-c is an effective target for treating cancer-induced bone pain through the induction of AMPK-mediated mitochondrial biogenesis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1323-1339. [PMID: 38716540 PMCID: PMC11532206 DOI: 10.3724/abbs.2024048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/20/2024] [Indexed: 10/17/2024] Open
Abstract
Bone cancer pain (BCP), due to cancer bone metastasis and bone destruction, is a common symptom of tumors, including breast, prostate, and lung tumors. Patients often experience severe pain without effective treatment. Here, using a mouse model of bone cancer, we report that MOTS-c, a novel mitochondrial-derived peptide, confers remarkable protection against cancer pain and bone destruction. Briefly, we find that the plasma level of endogenous MOTS-c is significantly lower in the BCP group than in the sham group. Accordingly, intraperitoneal administration of MOTS-c robustly attenuates bone cancer-induced pain. These effects are blocked by compound C, an AMPK inhibitor. Furthermore, MOTS-c treatment significantly enhances AMPKα 1/2 phosphorylation. Interestingly, mechanical studies indicate that at the spinal cord level, MOTS-c relieves pain by restoring mitochondrial biogenesis, suppressing microglial activation, and decreasing the production of inflammatory factors, which directly contribute to neuronal modulation. However, in the periphery, MOTS-c protects against local bone destruction by modulating osteoclast and immune cell function in the tumor microenvironment, providing long-term relief from cancer pain. Additionally, we find that chronic administration of MOTS-c has little effect on liver, renal, lipid or cardiac function in mice. In conclusion, MOTS-c improves BCP through peripheral and central synergistic effects on nociceptors, immune cells, and osteoclasts, providing a pharmacological and biological rationale for the development of mitochondrial peptide-based therapeutic agents for cancer-induced pain.
Collapse
Affiliation(s)
- Long Yang
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Miaomiao Li
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Yucheng Liu
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| | - Yang Bai
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Tianyu Yin
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| | - Yangyang Chen
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| | - Jinhong Jiang
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
| | - Su Liu
- Jiangsu Province Key Laboratory of AnesthesiologyJiangsu Province Key Laboratory of Anesthesia and Analgesia Application TechnologyNMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic DrugsXuzhou Medical UniversityXuzhou221004China
- Department of Anesthesiologythe Affiliated Hospital of Xuzhou Medical UniversityXuzhou221018China
| |
Collapse
|
14
|
Wen Z, Fan J, Zhan F, Li X, Li B, Lu P, Yao X, Shen Z, Liu Z, Wang C, Li X, Jin W, Zhang X, Qi Y, Wang X, Song M. The role of FPR2-mediated ferroptosis in formyl peptide-induced acute lung injury against endothelial barrier damage and protective effect of the mitochondria-derived peptide MOTS-c. Int Immunopharmacol 2024; 131:111911. [PMID: 38527401 DOI: 10.1016/j.intimp.2024.111911] [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: 01/09/2024] [Revised: 02/19/2024] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Acute lung injury (ALI) has garnered significant attention in the field of respiratory and critical care due to its high mortality and morbidity, and limited treatment options. The role of the endothelial barrier in the development of ALI is crucial. Several bacterial pathogenic factors, including the bacteria-derived formyl peptide (fMLP), have been implicated in damaging the endothelial barrier and initiating ALI. However, the mechanism by which fMLP causes ALI remains unclear. In this study, we aim to explore the mechanisms of ALI caused by fMLP and evaluate the protective effects of MOTS-c, a mitochondrial-derived peptide. METHODS We established a rat model of ALI and a human pulmonary microvascular endothelial cell (HPMVEC) model of ALI by treatment with fMLP. In vivo experiments involved lung histopathology assays, assessments of inflammatory and oxidative stress factors, and measurements of ferroptosis-related proteins and barrier proteins to evaluate the severity of fMLP-induced ALI and the type of tissue damage in rats. In vitro experiments included evaluations of fMLP-induced damage on HPMVEC using cell activity assays, assessments of inflammatory and oxidative stress factors, measurements of ferroptosis-related proteins, endothelial barrier function assays, and examination of the key role of FPR2 in fMLP-induced ALI. We also assessed the protective effect of MOTS-c and investigated its mechanism on the fMLP-induced ALI in vivo and in vitro. RESULTS Results from both in vitro and in vivo experiments demonstrate that fMLP promotes the expression of inflammatory and oxidative stress factors, activates ferroptosis and disrupts the vascular endothelial barrier, ultimately contributing to the development and progression of ALI. Mechanistically, ferroptosis mediated by FPR2 plays a key role in fMLP-induced injury, and the Nrf2 and MAPK pathways are involved in this process. Knockdown of FPR2 and inhibition of ferroptosis can attenuate ALI induced by fMLP. Moreover, MOTS-c could protect the vascular endothelial barrier function by inhibiting ferroptosis and suppressing the expression of inflammatory and oxidative stress factors through Nrf2 and MAPK pathways, thereby alleviating fMLP-induced ALI. CONCLUSION Overall, fMLP disrupts the vascular endothelial barrier through FPR2-mediated ferroptosis, leading to the development and progression of ALI. MOTS-c demonstrates potential as a protective treatment against ALI by alleviating the damage induced by fMLP.
Collapse
Affiliation(s)
- Ziang Wen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Jidan Fan
- Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Faliang Zhan
- Department of Cardiothoracic Surgery, Yili Friendship Hospital, Yining, Xinjiang Uyghur Autonomous Region 839300, China
| | - Xiaopei Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ben Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Peng Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xin Yao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zihao Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zhaoyang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Chufan Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiangyu Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Wanjun Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiao Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanpu Qi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China; Department of Cardiovascular Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu 225300, China
| | - Meijuan Song
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| |
Collapse
|
15
|
Lin C, Luo L, Xun Z, Zhu C, Huang Y, Ye Y, Zhang J, Chen T, Wu S, Zhan F, Yang B, Liu C, Ran N, Ou Q. Novel function of MOTS-c in mitochondrial remodelling contributes to its antiviral role during HBV infection. Gut 2024; 73:338-349. [PMID: 37788894 DOI: 10.1136/gutjnl-2023-330389] [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: 05/31/2023] [Accepted: 09/16/2023] [Indexed: 10/05/2023]
Abstract
OBJECTIVE Hepatitis B virus (HBV) infection causes substantial harm to mitochondrial activity, which hinders the development of effective treatments for chronic hepatitis B (CHB). The discovery of the mitochondrial-derived short peptide MOTS-c, which possesses multiple bioactivities, offers a promising new approach in treating HBV infection. This study aims to explore the diagnostic and therapeutic potential of MOTS-c in HBV-related diseases and its molecular mechanism. DESIGN In total, 85 healthy subjects and 404 patients with HBV infection, including 20 clinical treatment cohorts, were recruited for this study. MOTS-c levels were measured by ELISA and its diagnostic value was evaluated by receiving operating characteristic curve analysis. The therapeutic effect of MOTS-c was observed in multiple HBV-infected mice and cells through various techniques, including transcriptomic sequencing, flow cytometry, immunofluorescence and electron microscopy. Additionally, MOTS-c's potential interaction with myosin-9 (MYH9) and actin was predicted using immunoprecipitation, proteomics and target prediction software. RESULTS MOTS-c negatively correlates with HBV DNA expression (R=-0.71), and its AUC (the area under the curve) for distinguishing CHB from healthy controls is 0.9530, and IA (immune reactive) from IC (inactive HBV carrier) is 0.8689. Inhibition of HBV replication (with a 50-70% inhibition rate) was observed alongside improved liver function without notable toxicity in vitro or in vivo. MOTS-c was found to promote mitochondrial biogenesis and enhance the MAVS (mitochondrial antiviral signalling protein) signalling pathway. The impact is dependent on MOTS-c's ability to regulate MYH9-actin-mediated mitochondrial homeostasis. CONCLUSION MOTS-c has the potential to serve as a biomarker for the progression of HBV infection while also enhancing antiviral efficacy. These findings present a promising innovative approach for effectively treating patients with CHB. Furthermore, our research uncovers a novel role for MOTS-c in regulating MYH9-actin-mediated mitochondrial dynamics and contributing to mitochondrial biogenesis.
Collapse
Affiliation(s)
- Caorui Lin
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Linjie Luo
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen Xun
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Chenggong Zhu
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Huang
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuchen Ye
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiawei Zhang
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Tianbin Chen
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Songhang Wu
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Fuguo Zhan
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Bin Yang
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Can Liu
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Ning Ran
- Institute of Medical Sciences, The Second Hospital & Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qishui Ou
- Department of Laboratory Medicine, Fujian Key Laboratory of Laboratory Medicine, Fujian Clinical Research Center for Clinical Immunology Laboratory Test, Gene Diagnosis Research Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Laboratory Medicine, National Reginal Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
16
|
Sahgal A, Uversky V, Davé V. Microproteins transitioning into a new Phase: Defining the undefined. Methods 2023; 220:38-54. [PMID: 37890707 DOI: 10.1016/j.ymeth.2023.10.009] [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: 09/01/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Recent advancements in omics technologies have unveiled a hitherto unknown group of short polypeptides called microproteins (miPs). Despite their size, accumulating evidence has demonstrated that miPs exert varied and potent biological functions. They act in paracrine, juxtracrine, and endocrine fashion, maintaining cellular physiology and driving diseases. The present study focuses on biochemical and biophysical analysis and characterization of twenty-four human miPs using distinct computational methods, including RIDAO, AlphaFold2, D2P2, FuzDrop, STRING, and Emboss Pep wheel. miPs often lack well-defined tertiary structures and may harbor intrinsically disordered regions (IDRs) that play pivotal roles in cellular functions. Our analyses define the physicochemical properties of an essential subset of miPs, elucidating their structural characteristics and demonstrating their propensity for driving or participating in liquid-liquid phase separation (LLPS) and intracellular condensate formation. Notably, miPs such as NoBody and pTUNAR revealed a high propensity for LLPS, implicating their potential involvement in forming membrane-less organelles (MLOs) during intracellular LLPS and condensate formation. The results of our study indicate that miPs have functionally profound implications in cellular compartmentalization and signaling processes essential for regulating normal cellular functions. Taken together, our methodological approach explains and highlights the biological importance of these miPs, providing a deeper understanding of the unusual structural landscape and functionality of these newly defined small proteins. Understanding their functions and biological behavior will aid in developing targeted therapies for diseases that involve miPs.
Collapse
Affiliation(s)
- Aayushi Sahgal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Biotechnology Graduate Program, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Vladimir Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Vrushank Davé
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Biotechnology Graduate Program, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Department of Pathology and Cell Biology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States; Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, United States.
| |
Collapse
|
17
|
Luo YH, Xie L, Li JY, Xie Y, Li MQ, Zhou L. Serum MOTS-C Levels are Decreased in Obese Children and Associated with Vascular Endothelial Function. Diabetes Metab Syndr Obes 2023; 16:1013-1020. [PMID: 37077579 PMCID: PMC10106799 DOI: 10.2147/dmso.s403934] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/28/2023] [Indexed: 04/20/2023] Open
Abstract
PURPOSE The increasing prevalence of obesity in children and its associated risk with cardiovascular diseases demand more discovery of the novel biomarkers for developing new treatment options for this complex disease. This study aimed to investigate the association of serum MOTS-C (a peptide encoded in the mitochondrial genome) levels and vascular endothelial function in obese children. PATIENTS AND METHODS A total of 225 obese children (aged 8.1 ± 2.6 years) and 218 healthy children (aged 7.9 ± 2.2 years) were enrolled. Related anthropometric assessment and biochemical evaluation were done in all subjects. Reactive hyperemia index (RHI), as assessed by the peripheral arterial tonometry, was used for evaluation of peripheral endothelial function. Enzyme-linked immunosorbent assay (ELISA) was used to measure the level of serum MOTS-C. RESULTS Levels of serum MOTS-C and RHI were lower in the obese children compared with the healthy children (P < 0.01). The RHI level was independently associated with body mass index, high-density lipoprotein cholesterol, and MOTS-C in linear regression analysis. Further analysis showed a significant mediating effect of MOTS-C on the correlation between body mass index and RHI in children, with the ratio of mediating effect value of 9.12%. CONCLUSION These data identify that MOTS-C is a previously unknown regulator in the development process of obesity-induced vascular changes.
Collapse
Affiliation(s)
- Yan-Hua Luo
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Li Xie
- Pediatric Medical Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Jiao-Yang Li
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Yuan Xie
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Man-Qin Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Li Zhou
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
- Correspondence: Li Zhou, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China, Tel +86 18573477875, Email
| |
Collapse
|