1
|
El-Tanbouly GS. Experimental verification of Myriocin molecular bioactivity in revoking rheumatoid arthritis: New emphasis on S100A1 repression and PPAR-γ activation via non-canonical pathways in the flare-up of cytokines. Int Immunopharmacol 2025; 158:114811. [PMID: 40354710 DOI: 10.1016/j.intimp.2025.114811] [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: 01/12/2025] [Revised: 04/19/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Myriocin (MYR), the immunosuppressive agent isolated from Mycelia sterilia is recently nominated as a potential therapy for various inflammatory disorders. This work is a primary insight on the mechanistic pathways for its potential anti-arthritic effect against complete Freund's adjuvant-induced arthritis in mice. First day double injection of 0.1 ml adjuvant, followed by a single booster dose in the second day provoked arthritis after 5 weeks. Initiation of treatment with MYR (0.4 mg/kg, i.p.) and the anti-arthritic drug; methotrexate (MT, 0.75 mg/kg, i.p., 3 times/week) was commenced from day 10 to the end of the study. MYR, similar to MT, mitigate arthritis progression through distinct lowering in serum rheumatoid factor, arthritis index, paw thickness, and lessen joint inflammation and destruction in limb architecture. MYR inhibited S100A1/TLR4 signaling, thus, reduced IL-23 generation and subsequent IL-17 production. MYR exerted additional anti-inflammatory effects via marked reduction in NF-κBp65, TNF-α, IL-1β, IL-6, and COX-2 tissue levels. These effects are linked with myriocin-induced PPAR-γ stimulatory actions, thus explaining the transrepression between PPAR-γ, S100A1/TLR4/NF-κB and IL-23/IL-17 signaling, with emphasizing the central role of PPAR-γ activation in the connection between multiple pathways. In view of these results, myriocin could be a possible innovative cure for immune/inflammatory disorders as rheumatoid arthritis.
Collapse
Affiliation(s)
- Ghada S El-Tanbouly
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| |
Collapse
|
2
|
Kourakis S, Timpani CA, Bagaric RM, Qi B, Ali BA, Boyer R, Spiesberger G, Kandhari N, Yan X, Kuang J, Tulangekar A, de Haan JB, Deveson-Lucas D, Stupka N, Fischer D, Rybalka E. Repurposed Nrf2 activator dimethyl fumarate rescues muscle inflammation and fibrosis in an aggravated mdx mouse model of Duchenne muscular dystrophy. Redox Biol 2025; 84:103676. [PMID: 40381228 DOI: 10.1016/j.redox.2025.103676] [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: 03/18/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Abstract
In inherited neuromuscular disease, Duchenne muscular dystrophy (DMD), glucocorticoids significantly slow disease progression yet impart side effects severe enough to preclude use in a significant proportion of patients. Extending our findings that acute treatment with FDA approved multiple sclerosis drug, dimethyl fumarate (DMF), rescues muscle pathology in juvenile mdx mice, we aimed to conduct tiered pre-clinical testing toward translation. To aggravate disease phenotype in adult mdx muscles that usually lack human equivalent muscle pathology, we used bi-weekly treadmill running for 4 weeks which increased plasma DMD biomarker, creatine kinase, by 2-fold and quadriceps fibrosis by ∼30 %. Using this model, we screened DMF for 5 weeks in a head-to-head comparison, and in combination, with standard-of-care prednisone (PRED), to model the most likely clinical trial scenario. We show comparable efficacy between DMF and PRED at reducing inflammation via NF-κB suppression and CD68+ macrophage infiltration. Moderate term DMF monotherapy had additional anti-fibrotic and anti-lipogenic effects on skeletal and cardiac muscle beyond those seen with PRED treatment, although combination therapy exacerbated fibrosis in quadriceps. Our study supports DMF as a repurposing candidate for DMD, especially for patients who cannot tolerate chronic glucocorticoid treatment. We also highlight the importance of evaluating combination therapy to identify potential off-target effects between emerging therapeutics and glucocorticoids towards better designed clinical trials.
Collapse
Affiliation(s)
- Stephanie Kourakis
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Cara A Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia; Department of Medicine - Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Ryan M Bagaric
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Bo Qi
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Benazir A Ali
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Rebecca Boyer
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
| | - Guinevere Spiesberger
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Nitika Kandhari
- Monash Genomics and Bioinformatics Platform, Biomedical Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Xu Yan
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Sarcopenia Research Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Jujiao Kuang
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
| | - Ankita Tulangekar
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Judy B de Haan
- Basic Science Domain, Oxidative Stress Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Deanna Deveson-Lucas
- Monash Genomics and Bioinformatics Platform, Biomedical Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Nicole Stupka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Department of Medicine - Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Dirk Fischer
- Division of Neuropediatric and Developmental Medicine, University Children's Hospital of Basel (UKBB), Basel, Switzerland
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia; Inherited and Acquired Myopathies Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia.
| |
Collapse
|
3
|
Wang J, Shan J, Guo C, Duan Y, Zhang F, Ye W, Liu Y. Transcriptome analysis and machine learning methods reveal potential mechanisms of zebrafish muscle aging. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101532. [PMID: 40367591 DOI: 10.1016/j.cbd.2025.101532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
Muscle is one of the most abundant tissues in the human body, and its aging usually leads to many adverse consequences. Zebrafish is a powerful model used to study human muscle diseases, yet we know little about the molecular mechanisms of muscle aging in zebrafish. In this study, we determined the gene expression profiles of muscle tissues from male zebrafish of four different ages. Through differential expression analysis and expression pattern analysis, we identified a set of genes associated with muscle aging in zebrafish. Functional enrichment analysis revealed that several biological changes accompanied zebrafish muscle aging, including chronic inflammation, accumulation of sphingolipids, reduction of autophagy, and activation of the ferroptosis pathway. H&E staining showed that zebrafish muscle senescence leads to myofibrillar interstitial expansion and inflammatory cell infiltration. Furthermore, we screened zebrafish muscle aging related biomarkers by machine learning and verified the expression levels of some biomarkers by RT-qPCR. Based on these biomarkers, we constructed a zebrafish muscle aging clock that can predict muscle age based on transcriptomic data. This study provides us with a new perspective to understand the molecular mechanism of muscle aging and a new tool for zebrafish-based anti-aging research.
Collapse
Affiliation(s)
- Jian Wang
- Anesthesiology Department, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, Zhejiang, China
| | - Junwei Shan
- Hunter Biotechnology, Inc., Hangzhou 310051, China
| | - Cheng Guo
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China; Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - You Duan
- West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Zhang
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| | - Weidong Ye
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| | - Yanxiao Liu
- Department of Spine Surgery, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| |
Collapse
|
4
|
Niasse‐Sy Z, Zhao B, Lenardič A, Luong HTT, Bar‐Nur O, Auwerx J, Wohlwend M. Delivery of A Chemically Modified Noncoding RNA Domain Improves Dystrophic Myotube Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410908. [PMID: 39960339 PMCID: PMC12120708 DOI: 10.1002/advs.202410908] [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] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/28/2025] [Indexed: 05/31/2025]
Abstract
Fast twitch muscle fibers are prone to degradation in skeletal muscle pathologies, such as sarcopenia and muscular dystrophies. We previously showed that the exercise-induced long noncoding RNA CYTOR promotes fast-twitch myogenesis. Here, we identify an independent functional element within human CYTOR, and optimize its RNA delivery. In human primary myoblasts exogenous CYTOR exon 2 recapitulates the effect of full-length CYTOR by boosting fast-twitch myogenic differentiation. Furthermore, chemically modified CYTOR exon 2 RNAΨU (N1-me-PseudoU, 7-methyl guanosine 5'Cap, polyA) enhances RNA stability and reduces immunogenicity to CYTORexon2 RNA. Viral- or chemically optimized RNA-mediated CYTORexon2 administration drives commitment toward myogenic maturation in Duchenne muscular dystrophy-derived primary myoblasts, myogenic progenitor cells, and mouse embryonic stem cells. Furthermore, CYTORexon2, m1ΨU improves key disease characteristics in dystrophic myotubes, including calcium handling and mitochondrial bioenergetics. In summary, we identify CYTOR exon 2 as the functional domain of CYTOR that can be delivered in a disease context using chemical modifications. This is of particular importance given the susceptibility of fast muscle fibers in different muscle pathologies such as aging and dystrophies, and the oncogenic effect of CYTOR exon 1. This study, therefore, highlights the potential of identifying functional domains in noncoding RNAs. Delivery, or targeting of RNA domains might constitute next-generation RNA therapeutics.
Collapse
Affiliation(s)
- Zeinabou Niasse‐Sy
- Laboratory of Integrative Systems PhysiologyÉcole polytechnique fédérale de Lausanne (EPFL)Lausanne1015Switzerland
- Université LyonFaculté de Médecine et de Maïeutique Lyon‐Sud Charles MerieuxOullins69007France
- Hospices Civils de LyonGroupement Hospitalier SudService de GériatrieLyon69002France
| | - Bo Zhao
- Massachusetts Institute of Technology (MIT)Cambridge02139USA
| | - Ajda Lenardič
- Laboratory of Regenerative and Muscle BiologyInstitute of Human Movement Sciences and SportDepartment of Health Sciences and TechnologyETH ZurichSchwerzenbach8603Switzerland
| | - Huyen Thuc Tran Luong
- Laboratory of Integrative Systems PhysiologyÉcole polytechnique fédérale de Lausanne (EPFL)Lausanne1015Switzerland
| | - Ori Bar‐Nur
- Laboratory of Regenerative and Muscle BiologyInstitute of Human Movement Sciences and SportDepartment of Health Sciences and TechnologyETH ZurichSchwerzenbach8603Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems PhysiologyÉcole polytechnique fédérale de Lausanne (EPFL)Lausanne1015Switzerland
| | - Martin Wohlwend
- Massachusetts Institute of Technology (MIT)Cambridge02139USA
| |
Collapse
|
5
|
Tammineni ER, Manno C, Oza G, Figueroa L. Skeletal muscle disorders as risk factors for type 2 diabetes. Mol Cell Endocrinol 2025; 599:112466. [PMID: 39848431 PMCID: PMC11886953 DOI: 10.1016/j.mce.2025.112466] [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: 11/12/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The incidence and prevalence of muscular disorders and of type 2 diabetes (T2D) is increasing and both represent highly significant healthcare problems, both economically and compromising quality of life. Interestingly, skeletal muscle dysfunction and T2D share some commonalities including dysregulated glucose homeostasis, increased oxidative stress, dyslipidemia, and cytokine alterations. Several lines of evidence have hinted to a relationship between skeletal muscle dysfunction and T2D. For instance, T2D affects skeletal muscle morphology, functionality, and overall health through altered protein metabolism, impaired mitochondrial function, and ultimately cell viability. Conversely, humans suffering from myopathies and their experimental models demonstrated increased incidence of T2D through altered muscle glucose disposal function due to abnormal calcium homeostasis, compromised mitochondrial function, dyslipidemia, increased inflammatory cytokines and fiber size alterations and disproportions. Lifestyle modifications are essential for improving and maintaining mobility and metabolic health in individuals suffering from myopathies along with T2D. In this review, we updated current literature evidence on clinical incidence of T2D in inflammatory, mitochondrial, metabolic myopathies, and muscular dystrophies and further discussed the molecular basis of these skeletal muscle disorders leading to T2D.
Collapse
Affiliation(s)
| | - Carlo Manno
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| | - Goldie Oza
- Centro de Investigación y Desarrollo Tecnológico en Electroquímica S. C., Queretaro, Mexico
| | - Lourdes Figueroa
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| |
Collapse
|
6
|
Jeon EY, Kwak Y, Kang H, Kim H, Jin SY, Park S, Kim RG, Ko D, Won JK, Cho A, Jung I, Lee CH, Park J, Kim HY, Chae JH, Choi M. Inhibiting EZH2 complements steroid effects in Duchenne muscular dystrophy. SCIENCE ADVANCES 2025; 11:eadr4443. [PMID: 40085707 PMCID: PMC11908487 DOI: 10.1126/sciadv.adr4443] [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: 07/02/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating X-linked disorder caused by dystrophin gene mutations. Despite recent advances in understanding the disease etiology and applying emerging treatment methodologies, glucocorticoid derivatives remain the only general therapeutic option that can slow disease development. However, the precise molecular mechanism of glucocorticoid action remains unclear, and there is still need for additional remedies to complement the treatment. Here, using single-nucleus RNA sequencing and spatial transcriptome analyses of human and mouse muscles, we investigated pathogenic features in patients with DMD and palliative effects of glucocorticoids. Our approach further illuminated the importance of proliferating satellite cells and revealed increased activity of a signal transduction pathway involving EZH2 in the patient cells. Subsequent administration of EZH2 inhibitors to Dmd mutant mice resulted in improved muscle phenotype through maintaining the immune-suppressing effect but overriding the muscle weakness and fibrogenic effects exerted by glucocorticoids. Our analysis reveals pathogenic mechanisms that can be readily targeted by extant therapeutic options for DMD.
Collapse
MESH Headings
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Animals
- Humans
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/genetics
- Mice
- Glucocorticoids/pharmacology
- Glucocorticoids/therapeutic use
- Male
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Disease Models, Animal
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/drug effects
- Signal Transduction/drug effects
- Steroids/pharmacology
Collapse
Affiliation(s)
- Eun Young Jeon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yejin Kwak
- Department of Information Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
| | - Hyeji Kang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hanbyeol Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Se Young Jin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Soojin Park
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ryeo Gyeong Kim
- Department of Pediatrics, Rare Disease Center, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Dayoung Ko
- Department of Pediatric Surgery, Seoul National University Children’s Hospital, Seoul, Republic of Korea
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Anna Cho
- Department of Pediatrics, Rare Disease Center, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Inkyung Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Chul-Hwan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeongbin Park
- Department of Information Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
| | - Hyun-Young Kim
- Department of Pediatric Surgery, Seoul National University Children’s Hospital, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
7
|
Poisson J, Daskalaki I, Potluri V, Morel JD, Rodriguez-Lopez S, De Masi A, Benegiamo G, Jain S, Lima T, Auwerx J. Safe and Orally Bioavailable Inhibitor of Serine Palmitoyltransferase Improves Age-Related Sarcopenia. ACS Pharmacol Transl Sci 2025; 8:203-215. [PMID: 39816804 PMCID: PMC11729425 DOI: 10.1021/acsptsci.4c00587] [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: 10/07/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025]
Abstract
The accumulation of ceramides and related metabolites has emerged as a pivotal mechanism contributing to the onset of age-related diseases. However, small molecule inhibitors targeting the ceramide de novo synthesis pathway for clinical use are currently unavailable. We synthesized a safe and orally bioavailable inhibitor, termed ALT-007, targeting the rate-limiting enzyme of ceramide de novo synthesis, serine palmitoyltransferase (SPT). In a mouse model of age-related sarcopenia, ALT-007, administered through the diet, effectively restored muscle mass and function compromised by aging. Mechanistic studies revealed that ALT-007 enhances protein homeostasis in Caenorhabditis elegans and mouse models of aging and age-related diseases, such as sarcopenia and inclusion body myositis (IBM); this effect is mediated by a specific reduction in very-long chain 1-deoxy-sphingolipid species, which accumulate in both muscle and brain tissues of aged mice and in muscle cells from IBM patients. These findings unveil a promising therapeutic avenue for developing safe ceramide inhibitors to address age-related neuromuscular diseases.
Collapse
Affiliation(s)
- Johanne Poisson
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Ioanna Daskalaki
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Vijay Potluri
- Intonation
Research Laboratories, Hyderabad 500076, India
| | - Jean-David Morel
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Sandra Rodriguez-Lopez
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Alessia De Masi
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Giorgia Benegiamo
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Suresh Jain
- Intonation
Research Laboratories, Hyderabad 500076, India
| | - Tanes Lima
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Johan Auwerx
- Laboratory
of Integrative Systems Physiology, École
Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| |
Collapse
|
8
|
Wijekoon N, Gonawala L, Ratnayake P, Sirisena D, Gunasekara H, Dissanayake A, Amaratunga D, Steinbusch HWM, Hathout Y, Hoffman EP, Dalal A, Mohan C, de Silva KRD. Serum metabolomic signatures of patients with rare neurogenetic diseases: an insight into potential biomarkers and treatment targets. Front Mol Neurosci 2025; 17:1482999. [PMID: 39866907 PMCID: PMC11759312 DOI: 10.3389/fnmol.2024.1482999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction To further advance our understanding of Muscular Dystrophies (MDs) and Spinocerebellar Ataxias (SCAs), it is necessary to identify the biological patterns associated with disease pathology. Although progress has been made in the fields of genetics and transcriptomics, there is a need for proteomics and metabolomics studies. The present study aimed to be the first to document serum metabolic signatures of MDs (DMD, BMD, and LGMD 2A) SCAs (SCA 1-3), from a South Asian perspective. Methods A total of 28 patients (SCA 1-10, SCA 2-2, SCA 3-2, DMD-10, BMD-2, LGMD-2) and eight controls (aged 8-65 years) were included. Metabolomic analysis was performed by Ultrahigh Performance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS), with support from the Houston Omics Collaborative. Results and discussion Amino acid metabolism was the primary altered super pathway in DMD followed by carbohydrate metabolism and lipid metabolism. In contrast, BMD and LGMD 2A exhibited a more prominent alteration in lipid metabolism followed by amino acid metabolism. In SCAs, primarily lipid, amino acid, peptide, nucleotide, and xenobiotics pathways are affected. Our findings offer new insights into the variance of metabolite levels in MD and SCA, with substantial implications for pathology, drug development, therapeutic targets and clinical management. Intriguingly, this study identified two novel metabolites associated with SCA. This pilot cross-sectional study warrants further research involving larger groups of participants, to validate our findings.
Collapse
Affiliation(s)
- Nalaka Wijekoon
- Interdisciplinary Centre for Innovations in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Lakmal Gonawala
- Interdisciplinary Centre for Innovations in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | | | | | | | | | | | - Harry W. M. Steinbusch
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, United States
| | - Eric P. Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, United States
| | - Ashwin Dalal
- Diagnostics Division, Center for DNA Fingerprinting and Diagnostics, Hyderabad, India
| | - Chandra Mohan
- Department of Bioengineering, University of Houston, Houston, TX, United States
| | - K. Ranil D. de Silva
- Interdisciplinary Centre for Innovations in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
9
|
Liu J, Wang S, Shen Y, Shi H, Han L. Lipid metabolites and sarcopenia-related traits: a Mendelian randomization study. Diabetol Metab Syndr 2024; 16:231. [PMID: 39285470 PMCID: PMC11406728 DOI: 10.1186/s13098-024-01465-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE To explore the influence of lipid metabolism on the risk of sarcopenia. METHODS Two-sample Mendelian randomization (MR) analysis was used to determine causality. A total of 179 lipid metabolism data points were used for exposure, and the data were obtained from a plasma lipid metabolite study of 7174 participants. The total muscle mass and total muscle strength, as well as the muscle strength and muscle mass of different sex groups, were selected as the relevant traits of sarcopenia. Data for outcomes were obtained from the UK Biobank, and sample sizes ranged from 135 468 to 450 243. Inverse-variance weighted (IVW), as the main method for evaluating the causal relationship between lipid metabolites and sarcopenia, uses the false discovery rate (FDR) for multiple comparisons and conducts heterogeneity, pleiotropy, and reverse causality tests. RESULTS Twenty-seven lipid metabolites, mainly phosphatidylcholine, phosphatidylethanolamine, ceramide, triacylglycerol, sphingomyelin, and sterol ester, were found to be associated with the risk of sarcopenia. Ceramide (d40:1), ceramide (d40:2), and sterol ester are risk factors for decreased muscle mass and strength. There is a positive causal relationship between various phosphatidylcholine lipids and muscle mass and strength. Sphingomyelin (d42:2) is a protective factor for total muscle strength and female muscle strength. There are inconsistent effects between different lipid metabolites, triacylglycerol, and muscle strength and muscle mass. CONCLUSIONS There was a causal relationship between 27 lipid metabolites and sarcopenia traits, and targeting specific lipid metabolites may benefit sarcopenia diagnosis, disease assessment, and treatment.
Collapse
Affiliation(s)
- Jianping Liu
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Sufang Wang
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Yuan Shen
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Haicun Shi
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China
| | - Lijian Han
- Department of Neurology, Yancheng Third People's Hospital (The Sixth Affiliated Hospital of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, The affiliated hospital of Jiangsu Vocational College of Medicine), Yancheng, Jiangsu, China.
| |
Collapse
|
10
|
De Masi A, Zanou N, Strotjohann K, Lee D, Lima TI, Li X, Jeon J, Place N, Jung H, Auwerx J. Cyclo His-Pro Attenuates Muscle Degeneration in Murine Myopathy Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305927. [PMID: 38728626 PMCID: PMC11267275 DOI: 10.1002/advs.202305927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/11/2024] [Indexed: 05/12/2024]
Abstract
Among the inherited myopathies, a group of muscular disorders characterized by structural and metabolic impairments in skeletal muscle, Duchenne muscular dystrophy (DMD) stands out for its devastating progression. DMD pathogenesis is driven by the progressive degeneration of muscle fibers, resulting in inflammation and fibrosis that ultimately affect the overall muscle biomechanics. At the opposite end of the spectrum of muscle diseases, age-related sarcopenia is a common condition that affects an increasing proportion of the elderly. Although characterized by different pathological mechanisms, DMD and sarcopenia share the development of progressive muscle weakness and tissue inflammation. Here, the therapeutic effects of Cyclo Histidine-Proline (CHP) against DMD and sarcopenia are evaluated. In the mdx mouse model of DMD, it is shown that CHP restored muscle contractility and force production, accompanied by the reduction of fibrosis and inflammation in skeletal muscle. CHP furthermore prevented the development of cardiomyopathy and fibrosis in the diaphragm, the two leading causes of death for DMD patients. CHP also attenuated muscle atrophy and functional deterioration in a mouse model of age-related sarcopenia. These findings from two different models of muscle dysfunction hence warrant further investigation into the effects of CHP on muscle pathologies in animal models and eventually in patients.
Collapse
Affiliation(s)
- Alessia De Masi
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical SciencesFaculty of Biology‐MedicineUniversity of LausanneLausanne1015Switzerland
| | - Keno Strotjohann
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Dohyun Lee
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
| | - Tanes I. Lima
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Jongsu Jeon
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
| | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical SciencesFaculty of Biology‐MedicineUniversity of LausanneLausanne1015Switzerland
| | - Hoe‐Yune Jung
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
- School of Interdisciplinary Bioscience and BioengineeringPohang University of Science and Technology (POSTECH)Pohang37673South Korea
| | - Johan Auwerx
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| |
Collapse
|
11
|
Nitahara-Kasahara Y, Posadas-Herrera G, Hirai K, Oda Y, Snagu-Miyamoto N, Yamanashi Y, Okada T. Characterization of disease-specific alterations in metabolites and effects of mesenchymal stromal cells on dystrophic muscles. Front Cell Dev Biol 2024; 12:1363541. [PMID: 38946797 PMCID: PMC11211584 DOI: 10.3389/fcell.2024.1363541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/22/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) is a genetic disorder caused by mutations in the dystrophin-encoding gene that leads to muscle necrosis and degeneration with chronic inflammation during growth, resulting in progressive generalized weakness of the skeletal and cardiac muscles. We previously demonstrated the therapeutic effects of systemic administration of dental pulp mesenchymal stromal cells (DPSCs) in a DMD animal model. We showed preservation of long-term muscle function and slowing of disease progression. However, little is known regarding the effects of cell therapy on the metabolic abnormalities in DMD. Therefore, here, we aimed to investigate the mechanisms underlying the immunosuppressive effects of DPSCs and their influence on DMD metabolism. Methods A comprehensive metabolomics-based approach was employed, and an ingenuity pathway analysis was performed to identify dystrophy-specific metabolomic impairments in the mdx mice to assess the therapeutic response to our established systemic DPSC-mediated cell therapy approach. Results and Discussion We identified DMD-specific impairments in metabolites and their responses to systemic DPSC treatment. Our results demonstrate the feasibility of the metabolomics-based approach and provide insights into the therapeutic effects of DPSCs in DMD. Our findings could help to identify molecular marker targets for therapeutic intervention and predict long-term therapeutic efficacy.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Guillermo Posadas-Herrera
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kunio Hirai
- Division of Cell and Gene Therapy, Nippon Medical School, Tokyo, Japan
| | - Yuki Oda
- Division of Cell and Gene Therapy, Nippon Medical School, Tokyo, Japan
| | - Noriko Snagu-Miyamoto
- Division of Cell and Gene Therapy, Nippon Medical School, Tokyo, Japan
- Division of Oral and Maxillofacial Surgical, Tokyo Women’s Medical School, Tokyo, Japan
| | - Yuji Yamanashi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Wohlwend M, Laurila PP, Goeminne LJE, Lima T, Daskalaki I, Li X, von Alvensleben G, Crisol B, Mangione R, Gallart-Ayala H, Lalou A, Burri O, Butler S, Morris J, Turner N, Ivanisevic J, Auwerx J. Inhibition of CERS1 in skeletal muscle exacerbates age-related muscle dysfunction. eLife 2024; 12:RP90522. [PMID: 38506902 PMCID: PMC10954306 DOI: 10.7554/elife.90522] [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: 03/21/2024] Open
Abstract
Age-related muscle wasting and dysfunction render the elderly population vulnerable and incapacitated, while underlying mechanisms are poorly understood. Here, we implicate the CERS1 enzyme of the de novo sphingolipid synthesis pathway in the pathogenesis of age-related skeletal muscle impairment. In humans, CERS1 abundance declines with aging in skeletal muscle cells and, correlates with biological pathways involved in muscle function and myogenesis. Furthermore, CERS1 is upregulated during myogenic differentiation. Pharmacological or genetic inhibition of CERS1 in aged mice blunts myogenesis and deteriorates aged skeletal muscle mass and function, which is associated with the occurrence of morphological features typical of inflammation and fibrosis. Ablation of the CERS1 orthologue lagr-1 in Caenorhabditis elegans similarly exacerbates the age-associated decline in muscle function and integrity. We discover genetic variants reducing CERS1 expression in human skeletal muscle and Mendelian randomization analysis in the UK biobank cohort shows that these variants reduce muscle grip strength and overall health. In summary, our findings link age-related impairments in muscle function to a reduction in CERS1, thereby underlining the importance of the sphingolipid biosynthesis pathway in age-related muscle homeostasis.
Collapse
Affiliation(s)
- Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Ludger JE Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Tanes Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Ioanna Daskalaki
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Giacomo von Alvensleben
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Renata Mangione
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL)LausanneSwitzerland
| | - Amélia Lalou
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Olivier Burri
- Bioimaging and optics platform, École polytechnique fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Stephen Butler
- School of Chemistry, University of New South Wales SydneySydneyAustralia
| | - Jonathan Morris
- School of Chemistry, University of New South Wales SydneySydneyAustralia
| | - Nigel Turner
- Cellular Bioenergetics Laboratory, Victor Chang Cardiac Research InstituteDarlinghurstAustralia
- School of Biomedical Sciences, University of New South Wales SydneySydneyAustralia
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL)LausanneSwitzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de LausanneLausanneSwitzerland
| |
Collapse
|
13
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement. ACS Chem Neurosci 2024; 15:1-30. [PMID: 38095562 PMCID: PMC10767750 DOI: 10.1021/acschemneuro.3c00531] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/04/2024] Open
Abstract
Aging is a dynamic, time-dependent process that is characterized by a gradual accumulation of cell damage. Continual functional decline in the intrinsic ability of living organisms to accurately regulate homeostasis leads to increased susceptibility and vulnerability to diseases. Many efforts have been put forth to understand and prevent the effects of aging. Thus, the major cellular and molecular hallmarks of aging have been identified, and their relationships to age-related diseases and malfunctions have been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent aging-related research. We review the advances in knowledge and delineate trends in research advancements on aging factors and attributes across time and geography. We also review the current concepts related to the major aging hallmarks on the molecular, cellular, and organismic level, age-associated diseases, with attention to brain aging and brain health, as well as the major biochemical processes associated with aging. Major age-related diseases have been outlined, and their correlations with the major aging features and attributes are explored. We hope this review will be helpful for apprehending the current knowledge in the field of aging mechanisms and progression, in an effort to further solve the remaining challenges and fulfill its potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
14
|
Al Saedi A, Yacoub AS, Awad K, Karasik D, Brotto M, Duque G. The Interplay of Lipid Signaling in Musculoskeletal Cross Talk: Implications for Health and Disease. Methods Mol Biol 2024; 2816:1-11. [PMID: 38977583 DOI: 10.1007/978-1-0716-3902-3_1] [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: 07/10/2024]
Abstract
The intricate interplay between the muscle and bone tissues is a fundamental aspect of musculoskeletal physiology. Over the past decades, emerging research has highlighted the pivotal role of lipid signaling in mediating communication between these tissues. This chapter delves into the multifaceted mechanisms through which lipids, particularly phospholipids, sphingolipids, and eicosanoids, participate in orchestrating cellular responses and metabolic pathways in both muscle and bone. Additionally, we examine the clinical implications of disrupted lipid signaling in musculoskeletal disorders, offering insights into potential therapeutic avenues. This chapter aims to shed light on the complex lipid-driven interactions between the muscle and bone tissues, paving the way for a deeper understanding of musculoskeletal health and disease.
Collapse
Affiliation(s)
- Ahmed Al Saedi
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Ahmed S Yacoub
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - Kamal Awad
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - Gustavo Duque
- Research Institute of McGill University Health Center, Department of Medicine, McGill University, Québec, Canada
| |
Collapse
|
15
|
Chen Y, Hao T, Wang J, Chen Y, Wang X, Wei W, Zhao J, Qian Y. A Near-Infrared Fluorogenic Probe for Rapid, Specific, and Ultrasensitive Detection of Sphingosine in Living Cells and In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307598. [PMID: 38032131 PMCID: PMC10787105 DOI: 10.1002/advs.202307598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Indexed: 12/01/2023]
Abstract
Sphingosine (Sph) plays important roles in various complex biological processes. Abnormalities in Sph metabolism can result in various diseases, including neurodegenerative disorders. However, due to the lack of rapid and accurate detection methods, understanding sph metabolic in related diseases is limited. Herein, a series of near-infrared fluorogenic probes DMS-X (X = 2F, F, Cl, Br, and I) are designed and synthesized. The fast oxazolidinone ring formation enables the DMS-2F to detect Sph selectively and ultrasensitively, and the detection limit reaches 9.33 ± 0.41 nm. Moreover, it is demonstrated that DMS-2F exhibited a dose- and time-dependent response to Sph and can detect sph in living cells. Importantly, for the first time, the changes in Sph levels induced by Aβ42 oligomers and H2 O2 are assessed through a fluorescent imaging approach, and further validated the physiological processes by which Aβ42 oligomers and reactive oxygen species (ROS)-induce changes in intracellular Sph levels. Additionally, the distribution of Sph in living zebrafish is successfully mapped by in vivo imaging of a zebrafish model. This work provides a simple and efficient method for probing Sph in living cells and in vivo, which will facilitate investigation into the metabolic process of Sph and the connection between Sph and disease pathologies.
Collapse
Affiliation(s)
- Yanyan Chen
- State Key Laboratory of Coordination ChemistryChemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Tingting Hao
- State Key Laboratory of Coordination ChemistryChemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Jing Wang
- State Key Laboratory of Coordination ChemistryChemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yiming Chen
- School of EngineeringVanderbilt UniversityNashville37235USA
| | - Xiuxiu Wang
- State Key Laboratory of Coordination ChemistryChemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Wei Wei
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjing210023China
| | - Jing Zhao
- State Key Laboratory of Coordination ChemistryChemistry and Biomedicine Innovation Center (ChemBIC)School of Chemistry and Chemical EngineeringNanjing UniversityNanjing210023China
| | - Yong Qian
- Jiangsu Collaborative Innovation Center of Biomedical Functional MaterialsSchool of Chemistry and Materials ScienceNanjing Normal UniversityNanjing210023China
| |
Collapse
|
16
|
Vidal J, Fernandez EA, Wohlwend M, Laurila P, Lopez‐Mejia A, Ochala J, Lobrinus AJ, Kayser B, Lopez‐Mejia IC, Place N, Zanou N. Ryanodine receptor type 1 content decrease-induced endoplasmic reticulum stress is a hallmark of myopathies. J Cachexia Sarcopenia Muscle 2023; 14:2882-2897. [PMID: 37964752 PMCID: PMC10751419 DOI: 10.1002/jcsm.13349] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/24/2023] [Accepted: 09/11/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Decreased ryanodine receptor type 1 (RyR1) protein levels are a well-described feature of recessive RYR1-related myopathies. The aim of the present study was twofold: (1) to determine whether RyR1 content is also decreased in other myopathies and (2) to investigate the mechanisms by which decreased RyR1 protein triggers muscular disorders. METHODS We used publicly available datasets, muscles from human inflammatory and mitochondrial myopathies, an inducible muscle-specific RYR1 recessive mouse model and RyR1 knockdown in C2C12 muscle cells to measure RyR1 content and endoplasmic reticulum (ER) stress markers. Proteomics, lipidomics, molecular biology and transmission electron microscopy approaches were used to decipher the alterations associated with the reduction of RyR1 protein levels. RESULTS RYR1 transcripts were reduced in muscle samples of patients suffering from necrotizing myopathy (P = 0.026), inclusion body myopathy (P = 0.003), polymyositis (P < 0.001) and juvenile dermatomyositis (P < 0.001) and in muscle samples of myotonic dystrophy type 2 (P < 0.001), presymptomatic (P < 0.001) and symptomatic (P < 0.001) Duchenne muscular dystrophy, Becker muscular dystrophy (P = 0.004) and limb-girdle muscular dystrophy type 2A (P = 0.004). RyR1 protein content was also significantly decreased in inflammatory myopathy (-75%, P < 0.001) and mitochondrial myopathy (-71%, P < 0.001) muscles. Proteomics data showed that depletion of RyR1 protein in C2C12 myoblasts leads to myotubes recapitulating the common molecular alterations observed in myopathies. Mechanistically, RyR1 protein depletion reduces ER-mitochondria contact length (-26%, P < 0.001), Ca2+ transfer to mitochondria (-48%, P = 0.002) and the mitophagy gene Parkinson protein 2 transcripts (P = 0.037) and induces mitochondrial accumulation (+99%, P = 0.005) and dysfunction (P < 0.001). This was associated to the accumulation of deleterious sphingolipid species. Our data showed increased levels of the ER stress marker chaperone-binding protein/glucose regulated protein 78, GRP78-Bip, in RyR1 knockdown myotubes (+45%, P = 0.046), in mouse RyR1 recessive muscles (+58%, P = 0.001) and in human inflammatory (+96%, P = 0.006) and mitochondrial (+64%, P = 0.049) myopathy muscles. This was accompanied by increased protein levels of the pro-apoptotic protein CCAAT-enhancer-binding protein homologous protein, CHOP-DDIT3, in RyR1 knockdown myotubes (+27%, P < 0.001), mouse RyR1 recessive muscles (+63%, P = 0.009), human inflammatory (+50%, P = 0.038) and mitochondrial (+51%, P = 0.035) myopathy muscles. In publicly available datasets, the decrease in RYR1 content in myopathies was also associated to increased ER stress markers and RYR1 transcript levels are inversely correlated with ER stress markers in the control population. CONCLUSIONS Decreased RyR1 is commonly observed in myopathies and associated to ER stress in vitro, in mouse muscle and in human myopathy muscles, suggesting a potent role of RyR1 depletion-induced ER stress in the pathogenesis of myopathies.
Collapse
Affiliation(s)
- Jeremy Vidal
- Institute of Sport Sciences and Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
| | - Eric A. Fernandez
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Martin Wohlwend
- Computer Science and Artificial Intelligence LaboratoryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | - Andrea Lopez‐Mejia
- Center for Integrative GenomicsUniversity of LausanneLausanneSwitzerland
| | - Julien Ochala
- Department of Biomedical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Alexander J. Lobrinus
- Institute of PathologyLausanne University Hospital (CHUV)LausanneSwitzerland
- Department of Clinical PathologyUniversity Hospital GenevaGenevaSwitzerland
| | - Bengt Kayser
- Institute of Sport Sciences and Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
| | | | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical SciencesUniversity of LausanneLausanneSwitzerland
| |
Collapse
|
17
|
Rao H, Liu C, Wang A, Ma C, Xu Y, Ye T, Su W, Zhou P, Gao WQ, Li L, Ding X. SETD2 deficiency accelerates sphingomyelin accumulation and promotes the development of renal cancer. Nat Commun 2023; 14:7572. [PMID: 37989747 PMCID: PMC10663509 DOI: 10.1038/s41467-023-43378-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: 02/19/2023] [Accepted: 11/07/2023] [Indexed: 11/23/2023] Open
Abstract
Patients with polycystic kidney disease (PKD) encounter a high risk of clear cell renal cell carcinoma (ccRCC), a malignant tumor with dysregulated lipid metabolism. SET domain-containing 2 (SETD2) has been identified as an important tumor suppressor and an immunosuppressor in ccRCC. However, the role of SETD2 in ccRCC generation in PKD remains largely unexplored. Herein, we perform metabolomics, lipidomics, transcriptomics and proteomics within SETD2 loss induced PKD-ccRCC transition mouse model. Our analyses show that SETD2 loss causes extensive metabolic reprogramming events that eventually results in enhanced sphingomyelin biosynthesis and tumorigenesis. Clinical ccRCC patient specimens further confirm the abnormal metabolic reprogramming and sphingomyelin accumulation. Tumor symptom caused by Setd2 knockout is relieved by myriocin, a selective inhibitor of serine-palmitoyl-transferase and sphingomyelin biosynthesis. Our results reveal that SETD2 deficiency promotes large-scale metabolic reprogramming and sphingomyelin biosynthesis during PKD-ccRCC transition. This study introduces high-quality multi-omics resources and uncovers a regulatory mechanism of SETD2 on lipid metabolism during tumorigenesis.
Collapse
Affiliation(s)
- Hanyu Rao
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Changwei Liu
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Chunxiao Ma
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Xu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Tianbao Ye
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqiong Su
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Peijun Zhou
- Division of Kidney Transplant, Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Li Li
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
18
|
Xu H, Cai X, Xu K, Wu Q, Xu B. The metabolomic plasma profile of patients with Duchenne muscular dystrophy: providing new evidence for its pathogenesis. Orphanet J Rare Dis 2023; 18:273. [PMID: 37670327 PMCID: PMC10481483 DOI: 10.1186/s13023-023-02885-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 08/25/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a fatal genetic muscle-wasting disease that affects 1 in 5000 male births with no current cure. Despite great progress has been made in the research of DMD, its underlying pathological mechanism based on the metabolomics is still worthy of further study. Therefore, it is necessary to gain a deeper understanding of the mechanisms or pathogenesis underlying DMD, which may reveal potential therapeutic targets and/or biomarkers. RESULTS Plasma samples from 42 patients with DMD from a natural history study and 40 age-matched healthy volunteers were subjected to a liquid chromatography-mass spectrometry-based non-targeted metabolomics approach. Acquired metabolic data were evaluated by principal component analysis, partial least squares-discriminant analysis, and metabolic pathway analysis to explore distinctive metabolic patterns in patients with DMD. Differentially expressed metabolites were identified using publicly available and integrated databases. By comparing the DMD and healthy control groups, 25 differential metabolites were detected, including amino acids, unsaturated fatty acids, carnitine, lipids, and metabolites related to the gut microbiota. Correspondingly, linoleic acid metabolism, D-glutamine and D-glutamate metabolism, glycerophospholipid metabolism, and alanine, aspartate, and glutamate metabolism were significantly altered in patients with DMD, compared with those of healthy volunteers. CONCLUSIONS Our study demonstrated the abnormal metabolism of amino acids, energy, and lipids in patients with DMD, consistent with pathological features, such as recurrent muscle necrosis and regeneration, interstitial fibrosis, and fat replacement. Additionally, we found that metabolites of intestinal flora were disordered in DMD patients, providing support for treatment of intestinal microbia disturbance in DMD diseases. Our study provides a new research strategy for understanding the pathogenesis of DMD.
Collapse
Affiliation(s)
- Huayan Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaotang Cai
- Department of Rehabilitation Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qihong Wu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bei Xu
- Department of Clinical Laboratory, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, Sichuan, China.
- Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
19
|
Roy A, Koike TE, Joshi AS, Tomaz da Silva M, Mathukumalli K, Wu M, Kumar A. Targeted regulation of TAK1 counteracts dystrophinopathy in a DMD mouse model. JCI Insight 2023; 8:e164768. [PMID: 37071470 PMCID: PMC10322678 DOI: 10.1172/jci.insight.164768] [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: 08/24/2022] [Accepted: 04/12/2023] [Indexed: 04/19/2023] Open
Abstract
Muscular dystrophies make up a group of genetic neuromuscular disorders that involve severe muscle wasting. TGF-β-activated kinase 1 (TAK1) is an important signaling protein that regulates cell survival, growth, and inflammation. TAK1 has been recently found to promote myofiber growth in the skeletal muscle of adult mice. However, the role of TAK1 in muscle diseases remains poorly understood. In the present study, we have investigated how TAK1 affects the progression of dystrophic phenotype in the mdx mouse model of Duchenne muscular dystrophy (DMD). TAK1 is highly activated in the dystrophic muscle of mdx mice during the peak necrotic phase. While targeted inducible inactivation of TAK1 inhibits myofiber injury in young mdx mice, it results in reduced muscle mass and contractile function. TAK1 inactivation also causes loss of muscle mass in adult mdx mice. By contrast, forced activation of TAK1 through overexpression of TAK1 and TAB1 induces myofiber growth without having any deleterious effect on muscle histopathology. Collectively, our results suggest that TAK1 is a positive regulator of skeletal muscle mass and that targeted regulation of TAK1 can suppress myonecrosis and ameliorate disease progression in DMD.
Collapse
|
20
|
Lima TI, Laurila PP, Wohlwend M, Morel JD, Goeminne LJE, Li H, Romani M, Li X, Oh CM, Park D, Rodríguez-López S, Ivanisevic J, Gallart-Ayala H, Crisol B, Delort F, Batonnet-Pichon S, Silveira LR, Sankabattula Pavani Veera Venkata L, Padala AK, Jain S, Auwerx J. Inhibiting de novo ceramide synthesis restores mitochondrial and protein homeostasis in muscle aging. Sci Transl Med 2023; 15:eade6509. [PMID: 37196064 DOI: 10.1126/scitranslmed.ade6509] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 04/28/2023] [Indexed: 05/19/2023]
Abstract
Disruption of mitochondrial function and protein homeostasis plays a central role in aging. However, how these processes interact and what governs their failure in aging remain poorly understood. Here, we showed that ceramide biosynthesis controls the decline in mitochondrial and protein homeostasis during muscle aging. Analysis of transcriptome datasets derived from muscle biopsies obtained from both aged individuals and patients with a diverse range of muscle disorders revealed that changes in ceramide biosynthesis, as well as disturbances in mitochondrial and protein homeostasis pathways, are prevalent features in these conditions. By performing targeted lipidomics analyses, we found that ceramides accumulated in skeletal muscle with increasing age across Caenorhabditis elegans, mice, and humans. Inhibition of serine palmitoyltransferase (SPT), the rate-limiting enzyme of the ceramide de novo synthesis, by gene silencing or by treatment with myriocin restored proteostasis and mitochondrial function in human myoblasts, in C. elegans, and in the skeletal muscles of mice during aging. Restoration of these age-related processes improved health and life span in the nematode and muscle health and fitness in mice. Collectively, our data implicate pharmacological and genetic suppression of ceramide biosynthesis as potential therapeutic approaches to delay muscle aging and to manage related proteinopathies via mitochondrial and proteostasis remodeling.
Collapse
Affiliation(s)
- Tanes I Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Jean David Morel
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Ludger J E Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Hao Li
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Mario Romani
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, South Korea
| | - Dohyun Park
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Sandra Rodríguez-López
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne 1005, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne 1005, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Florence Delort
- Laboratoire Biologie Fonctionnelle et Adaptative, UMR 8251, CNRS and Université Paris Cité, Paris 8251, France
| | - Sabrina Batonnet-Pichon
- Laboratoire Biologie Fonctionnelle et Adaptative, UMR 8251, CNRS and Université Paris Cité, Paris 8251, France
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center, University of Campinas, Campinas 13083-864, Brazil
| | | | - Anil K Padala
- Intonation Research Laboratories, Hyderabad 500076, India
| | - Suresh Jain
- Intonation Research Laboratories, Hyderabad 500076, India
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| |
Collapse
|
21
|
Hepowit NL, Blalock E, Lee S, Bretland KM, MacGurn JA, Dickson RC. Reduced sphingolipid biosynthesis modulates proteostasis networks to enhance longevity. Aging (Albany NY) 2023; 15:472-491. [PMID: 36640272 PMCID: PMC9925692 DOI: 10.18632/aging.204485] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/29/2022] [Indexed: 01/15/2023]
Abstract
As the elderly population increases, chronic, age-associated diseases are challenging healthcare systems around the world. Nutrient limitation is well known to slow the aging process and improve health. Regrettably, practicing nutrient restriction to improve health is unachievable for most people. Alternatively, pharmacological strategies are being pursued including myriocin which increases lifespan in budding yeast. Myriocin impairs sphingolipid synthesis, resulting in lowered amino acid pools which promote entry into a quiescent, long-lived state. Here we present transcriptomic data during the first 6 hours of drug treatment that improves our mechanistic understanding of the cellular response to myriocin and reveals a new role for ubiquitin in longevity. Previously we found that the methionine transporter Mup1 traffics to the plasma membrane normally in myriocin-treated cells but is not active and undergoes endocytic clearance. We now show that UBI4, a gene encoding stressed-induced ubiquitin, is vital for myriocin-enhanced lifespan. Furthermore, we show that Mup1 fused to a deubiquitinase domain impairs myriocin-enhanced longevity. Broader effects of myriocin treatment on ubiquitination are indicated by our finding of a significant increase in K63-linked ubiquitin polymers following myriocin treatment. Although proteostasis is broadly accepted as a pillar of aging, our finding that ubiquitination of an amino acid transporter promotes longevity in myriocin-treated cells is novel. Addressing the role of ubiquitination/deubiquitination in longevity has the potential to reveal new strategies and targets for promoting healthy aging.
Collapse
Affiliation(s)
- Nathaniel L. Hepowit
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Eric Blalock
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY 40536, USA
| | - Sangderk Lee
- College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Kimberly M. Bretland
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, KY 40536, USA
| | - Jason A. MacGurn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
| | - Robert C. Dickson
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
22
|
Wang Y, Shyh-Chang N. Sphingolipids mediate lipotoxicity in muscular dystrophies. LIFE MEDICINE 2022; 1:273-275. [PMID: 39872745 PMCID: PMC11749356 DOI: 10.1093/lifemedi/lnac015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/25/2022] [Indexed: 01/30/2025]
Affiliation(s)
- Yuefan Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
23
|
Laurila PP, Wohlwend M, Imamura de Lima T, Luan P, Herzig S, Zanou N, Crisol B, Bou-Sleiman M, Porcu E, Gallart-Ayala H, Handzlik MK, Wang Q, Jain S, D'Amico D, Salonen M, Metallo CM, Kutalik Z, Eichmann TO, Place N, Ivanisevic J, Lahti J, Eriksson JG, Auwerx J. Sphingolipids accumulate in aged muscle, and their reduction counteracts sarcopenia. NATURE AGING 2022; 2:1159-1175. [PMID: 37118545 DOI: 10.1038/s43587-022-00309-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/07/2022] [Indexed: 04/30/2023]
Abstract
Age-related muscle dysfunction and sarcopenia are major causes of physical incapacitation in older adults and currently lack viable treatment strategies. Here we find that sphingolipids accumulate in mouse skeletal muscle upon aging and that both genetic and pharmacological inhibition of sphingolipid synthesis prevent age-related decline in muscle mass while enhancing strength and exercise capacity. Inhibition of sphingolipid synthesis confers increased myogenic potential and promotes protein synthesis. Within the sphingolipid pathway, we show that accumulation of dihydroceramides is the culprit disturbing myofibrillar homeostasis. The relevance of sphingolipid pathways in human aging is demonstrated in two cohorts, the UK Biobank and Helsinki Birth Cohort Study in which gene expression-reducing variants of SPTLC1 and DEGS1 are associated with improved and reduced fitness of older individuals, respectively. These findings identify sphingolipid synthesis inhibition as an attractive therapeutic strategy for age-related sarcopenia and co-occurring pathologies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sébastien Herzig
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maroun Bou-Sleiman
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eleonora Porcu
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Michal K Handzlik
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Qi Wang
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Suresh Jain
- Intonation Research Laboratories, Secunderabad, India
| | - Davide D'Amico
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Minna Salonen
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zoltan Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
| | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, National University Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
24
|
McCluskey G, Donaghy C, Morrison KE, McConville J, Duddy W, Duguez S. The Role of Sphingomyelin and Ceramide in Motor Neuron Diseases. J Pers Med 2022; 12:jpm12091418. [PMID: 36143200 PMCID: PMC9501626 DOI: 10.3390/jpm12091418] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), Spinal Bulbar Muscular Atrophy (SBMA), and Spinal Muscular Atrophy (SMA) are motor neuron diseases (MNDs) characterised by progressive motor neuron degeneration, weakness and muscular atrophy. Lipid dysregulation is well recognised in each of these conditions and occurs prior to neurodegeneration. Several lipid markers have been shown to predict prognosis in ALS. Sphingolipids are complex lipids enriched in the central nervous system and are integral to key cellular functions including membrane stability and signalling pathways, as well as being mediators of neuroinflammation and neurodegeneration. This review highlights the metabolism of sphingomyelin (SM), the most abundant sphingolipid, and of its metabolite ceramide, and its role in the pathophysiology of neurodegeneration, focusing on MNDs. We also review published lipidomic studies in MNDs. In the 13 studies of patients with ALS, 12 demonstrated upregulation of multiple SM species and 6 demonstrated upregulation of ceramides. SM species also correlated with markers of clinical progression in five of six studies. These data highlight the potential use of SM and ceramide as biomarkers in ALS. Finally, we review potential therapeutic strategies for targeting sphingolipid metabolism in neurodegeneration.
Collapse
Affiliation(s)
- Gavin McCluskey
- Personalised Medicine Center, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Department of Neurology, Altnagelvin Hospital, Derry, BT47 6SB, UK
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
| | - Colette Donaghy
- Department of Neurology, Altnagelvin Hospital, Derry, BT47 6SB, UK
| | - Karen E. Morrison
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Faculty of Medicine, Health & Life Sciences, Queen’s University, Belfast BT9 6AG, UK
| | - John McConville
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Department of Neurology, Ulster Hospital, Dundonald, Belfast BT16 1RH, UK
| | - William Duddy
- Personalised Medicine Center, School of Medicine, Ulster University, Derry BT47 6SB, UK
| | - Stephanie Duguez
- Personalised Medicine Center, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Correspondence:
| |
Collapse
|
25
|
Morroni J, Schirone L, Valenti V, Zwergel C, Riera CS, Valente S, Vecchio D, Schiavon S, Ragno R, Mai A, Sciarretta S, Lozanoska-Ochser B, Bouchè M. Inhibition of PKCθ Improves Dystrophic Heart Phenotype and Function in a Novel Model of DMD Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23042256. [PMID: 35216371 PMCID: PMC8880527 DOI: 10.3390/ijms23042256] [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: 01/03/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic cardiac muscle inflammation and subsequent fibrotic tissue deposition are key features in Duchenne Muscular Dystrophy (DMD). The treatment of choice for delaying DMD progression both in skeletal and cardiac muscle are corticosteroids, supporting the notion that chronic inflammation in the heart plays a pivotal role in fibrosis deposition and subsequent cardiac dysfunction. Nevertheless, considering the adverse effects associated with long-term corticosteroid treatments, there is a need for novel anti-inflammatory therapies. In this study, we used our recently described exercised mdx (ex mdx) mouse model characterised by accelerated heart pathology, and the specific PKCθ inhibitor Compound 20 (C20), to show that inhibition of this kinase leads to a significant reduction in the number of immune cells infiltrating the heart, as well as necrosis and fibrosis. Functionally, C20 treatment also prevented the reduction in left ventricle fractional shortening, which was typically observed in the vehicle-treated ex mdx mice. Based on these findings, we propose that PKCθ pharmacological inhibition could be an attractive therapeutic approach to treating dystrophic cardiomyopathy
Collapse
Affiliation(s)
- Jacopo Morroni
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
| | - Valentina Valenti
- Department of Cardiology, Ospedale Santa Maria Goretti, 04100 Latina, Italy;
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Carles Sánchez Riera
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Daniele Vecchio
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
| | - Sonia Schiavon
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
| | - Rino Ragno
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
| | - Marina Bouchè
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
- Correspondence:
| |
Collapse
|