1
|
Gouda MM, Elsharkawy ER, He Y, Li X. Importance of Advanced Detection Methodologies from Plant Cells to Human Microsystems Targeting Anticancer Applications. Int J Mol Sci 2025; 26:4691. [PMID: 40429833 PMCID: PMC12112733 DOI: 10.3390/ijms26104691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The growing global demand for phytochemicals as bioactive sources is prompting scientists to develop methods that link their sensory properties to their mechanisms of action in cancer treatment. Recent techniques for tracking the actions of small plant metabolites (SPMs) from single-cell plant sources to their molecular anticancer biomarkers could provide valuable insights in this field. Among the critical methods discussed in this review are the real-time tracking of cell components through stable isotope probing (Sis) and microspectroscopy, which has attracted the attention of biotechnologists. Additionally, the precise pathways required for studying new insights into functional materials are discussed, based on high-resolution and accurate technologies, which could aid their functional categorization. Notably, the molecules under study have recently garnered attention for their anticancer applications due to advancements in effective evaluation techniques that surpass traditional methods. In December 2020, the Food and Drug Administration (FDA) authorized 89 SPMs as safe anticancer natural molecules. In conclusion, by combining spatiotemporal techniques and SPMs' mechanisms, they could facilitate the development of more exceptional, bio-efficient materials.
Collapse
Affiliation(s)
- Mostafa M. Gouda
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China or (M.M.G.); (Y.H.)
- Department of Nutrition and Food Science, National Research Centre, Giza 12622, Egypt
| | - Eman R. Elsharkawy
- Center for Health Research, Northern Border University, Arar 73213, Saudi Arabia
| | - Yong He
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China or (M.M.G.); (Y.H.)
| | - Xiaoli Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China or (M.M.G.); (Y.H.)
| |
Collapse
|
2
|
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
|
3
|
Chawla T, Baskar D, Polavarapu K, Preethish-Kumar V, Nashi S, Vengalil S, Kulanthaivelu K, Saini J, Huddar A, Unnikrishnan G, Nandeesh BN, Nalini A. Muscle MRI Pattern in Dysferlinopathy and its Correlation with Dysferlin Gait. Ann Indian Acad Neurol 2025; 28:182-188. [PMID: 40079678 PMCID: PMC12049224 DOI: 10.4103/aian.aian_987_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Magnetic resonance imaging (MRI) in dysferlinopathy has consistently demonstrated a particular pattern of affliction. We aimed to study muscle MRI characteristics of lower limbs in limb girdle muscular dystrophy (LGMD)-R2 phenotypes and correlate them with the gait pattern. METHODS Forty genetically and/or biopsy-proven cases of dysferlinopathy underwent muscle MRI of the lower limbs. The pattern and extent of fatty infiltration and edema were recorded. Spearman's correlation analysis was used to find the correlation between muscle involvement and demographics, duration of illness, Muscular Dystrophy Functional Rating Scale (MDFRS), genotype, and gait pattern. RESULTS Mean age at onset and duration of illness at evaluation were 21.5 ± 6.3 years and 7.15 ± 4.95 years, respectively. Male: Female of patients was 2:1. Long head of biceps femoris was most severely involved with relative sparing of short head. Specific MRI pattern was noted based on phenotype, though no genotypic correlation was observed. Adductor magnus and semimembranosus were more severely involved in LGMD and proximodistal (PD) forms compared to Miyoshi muscular dystrophy type 1 phenotype. In addition, tibialis posterior and extensor hallucis longus were more severely involved in PD compared to MM and LGMD phenotypes. MDFRS mobility domain and duration of illness correlated with MRI findings. Gait pattern analysis revealed more severe involvement of flexor hallucis longus compared to extensor hallucis longus. CONCLUSIONS Muscle involvement differed based on the phenotype. Characteristic great toe extension in PD phenotype showed an imaging correlation with more severe involvement of flexor hallucis longus compared to extensor hallucis longus. Thus, imaging can be a potential biomarker to study the evolution and severity of disease in dysferlinopathy.
Collapse
Affiliation(s)
- Tanushree Chawla
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Dipti Baskar
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
- Department of Neurology, Children’s Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | | | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Karthik Kulanthaivelu
- Department of Neuro Imaging and Interventional Radiology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Jitender Saini
- Department of Neuro Imaging and Interventional Radiology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Akshata Huddar
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | - Gopikrishnan Unnikrishnan
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| | | | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, India
| |
Collapse
|
4
|
Ouellette JM, Mallender MD, Hian-Cheong DJ, Scurto DL, Nicholas JE, Trumble SJ, Hawke TJ, Krause MP. Altered sphingolipid profile in response to skeletal muscle injury in a mouse model of type 1 diabetes mellitus. Am J Physiol Cell Physiol 2025; 328:C273-C287. [PMID: 39611411 DOI: 10.1152/ajpcell.00158.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
A complication of type 1 diabetes mellitus (T1DM) is diabetic myopathy that includes reduced regenerative capacity of skeletal muscle. Sphingolipids are a diverse family of lipids with roles in skeletal muscle regeneration. Some studies have found changes in sphingolipid species levels in T1DM, however, the effect of T1DM on a sphingolipid panel in regenerating skeletal muscle has not been examined. Wild-type (WT) and diabetic Ins2Akita+/- (Akita) mice received cardiotoxin-induced muscle injury in their left quadriceps, gastrocnemius-plantaris-soleus, and tibialis anterior muscles with the contralateral muscles serving as uninjured controls. Muscles were collected at 1, 3, 5, or 7 days postinjury. In regenerating muscle from Akita mice, lipid staining with BODIPY 493/503 revealed increased intramyocellular and total lipids and perilipin-1-positive cell numbers as compared with WT. Liquid chromatography-mass spectrometry of quadriceps was used to identify sphingolipid levels in skeletal muscle. The C22:0 and C24:0 ceramides were significantly elevated in uninjured Akita, whereas ceramide C24:1 was decreased in injured Akita compared with WT. Ceramide-1-phosphate was increased in Akita compared with WT regardless of injury, whereas sphingosine-1-phosphate (S1P) was elevated with injury in WT but this response was muted in Akita mice. Western blotting of key enzymes involved in sphingolipid metabolism revealed S1P lyase, the enzyme that degrades S1P irreversibly, was significantly elevated in the injured muscle in Akita mice during regeneration, in accordance with lower S1P levels. This mouse model of T1DM demonstrates sphingolipidomic changes that may contribute to delayed muscle regeneration.NEW & NOTEWORTHY Muscle lipids become elevated, and the sphingolipid profile is altered by T1DM in skeletal muscle regeneration. A loss of S1P is accompanied by greater expression of sphingosine-1-phosphate lyase (SPL) in response to injury in Akita mice, suggesting a role for sphingolipids in the attenuated repair of skeletal muscle in T1DM rodent models. Although ceramide-1-phosphate (C1P) is increased with T1DM, there was no increase in ceramide kinase (CerK) suggesting an alternative route of ceramide phosphorylation in skeletal muscle.
Collapse
Affiliation(s)
- Jacob M Ouellette
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - Michael D Mallender
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - Dylan J Hian-Cheong
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - Daniel L Scurto
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | - James E Nicholas
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| | | | - Thomas J Hawke
- Department of Pathology & Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Matthew P Krause
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, Windsor, Ontario, Canada
| |
Collapse
|
5
|
Lloyd EM, Hepburn MS, Li J, Mowla A, Jeong JH, Hwang Y, Choi YS, Jackaman C, Kennedy BF, Grounds MD. Multimodal three-dimensional characterization of murine skeletal muscle micro-scale elasticity, structure, and composition: Impact of dysferlinopathy, Duchenne muscular dystrophy, and age on three hind-limb muscles. J Mech Behav Biomed Mater 2024; 160:106751. [PMID: 39326249 DOI: 10.1016/j.jmbbm.2024.106751] [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: 05/27/2024] [Revised: 08/21/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
Skeletal muscle tissue function is governed by the mechanical properties and organization of its components, including myofibers, extracellular matrix, and adipose tissue, which can be modified by the onset and progression of many disorders. This study used a novel combination of quantitative micro-elastography and clearing-enhanced three-dimensional (3D) microscopy to assess 3D micro-scale elasticity and micro-architecture of muscles from two muscular dystrophies: dysferlinopathy and Duchenne muscular dystrophy, using male BLA/J and mdx mice, respectively, and their wild-type (WT) controls. We examined three muscles with varying proportions of slow- and fast-twitch myofibers: the soleus (predominantly slow), extensor digitorum longus (EDL; fast), and quadriceps (mixed), from BLA/J and WTBLA/J mice aged 3, 10, and 24 months, and mdx and WTmdx mice aged 10 months. Both dysferlin deficiency and age reduced the elasticity and variability of elasticity of the soleus and quadriceps, but not EDL. Overall, the BLA/J soleus was 20% softer than WT and less mechanically heterogeneous (-14% in standard deviation of elasticity). The BLA/J quadriceps at 24 months was 72% softer than WT and less mechanically heterogeneous (-59% in standard deviation), with substantial adipose tissue accumulation. While mdx muscles did not differ quantitatively from WT, regional heterogeneity was evident in micro-scale elasticity and micro-architecture of quadriceps (e.g., 11.2 kPa in a region with marked pathology vs 3.8 kPa in a less affected area). These results demonstrate differing biomechanical changes in hind-limb muscles of two distinct muscular dystrophies, emphasizing the potential for this novel multimodal technique to identify important differences between various myopathies.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Curtin Health Innovation Research Institute, Curtin Medical School, Faculty of Health Sciences, Curtin University, Kent St, Bentley, Western Australia, 6102, Australia.
| | - Matt S Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland.
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.
| | - Alireza Mowla
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia.
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, 31151, Republic of Korea.
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan-si, Chungcheongnam-do, 31151, Republic of Korea.
| | - Yu Suk Choi
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia.
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin Medical School, Faculty of Health Sciences, Curtin University, Kent St, Bentley, Western Australia, 6102, Australia.
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia; Centre for Medical Research, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Electrical, Electronic & Computer Engineering, School of Engineering, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia; Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, 35 Stirling Highway, Perth, Western Australia, 6009, Australia.
| |
Collapse
|
6
|
Borland H, Moore U, Dressman HG, Human A, Mayhew AG, Hilsden H, Rufibach LE, Duong T, Maron E, DeWolf B, Rose K, Siener C, Thiele S, Práxedes NSA, Canal A, Holsten S, Sakamoto C, Pedrosa-Hernández I, Bello L, Alfano LN, Lowes LP, James MK, Straub V. Performance of upper limb entry item to predict forced vital capacity in dysferlin-deficient limb girdle muscular dystrophy. Neuromuscul Disord 2024; 43:20-28. [PMID: 39178649 DOI: 10.1016/j.nmd.2024.08.003] [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: 03/26/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
Dysferlin-deficient limb girdle muscular dystrophy (LGMD R2), also referred to as dysferlinopathy, can be associated with respiratory muscle weakness as the disease progresses. Clinical practice guidelines recommend biennial lung function assessments in patients with dysferlinopathy to screen for respiratory impairment. However, lack of universal access to spirometry equipment and trained specialists makes regular monitoring challenging. This study investigated the use of the Performance of Upper Limb (PUL) clinical scale entry item as a low-cost screening tool to identify patients with dysferlinopathy at risk of respiratory impairment. Using data from 193 patients from the Jain Foundation's International Clinical Outcomes Study, modelling identified a significant positive relationship between the PUL entry item and forced vital capacity (FVC). Eighty-eight percent of patients with the lowest PUL entry item score of 1 presented with FVC % predicted values of <60 %, suggestive of respiratory impairment. By contrast, only 10 % of the remainder of the cohort (PUL entry item of 2 or more) had an FVC of <60 %. This relationship also held true when accounting for ambulatory status, age, and sex as possible confounding factors. In summary, our results suggest that the PUL entry item could be implemented in clinical practice to screen for respiratory impairment where spirometry is not readily available.
Collapse
Affiliation(s)
- Holly Borland
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon, Tyne, United Kingdom
| | - Ursula Moore
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon, Tyne, United Kingdom
| | - Heather Gordish Dressman
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, United States; Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, DC, United States
| | - Anri Human
- University of Pretoria, Pretoria, South Africa
| | - Anna G Mayhew
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon, Tyne, United Kingdom
| | - Heather Hilsden
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon, Tyne, United Kingdom
| | | | - Tina Duong
- Stanford University, Palo Alto, United States
| | | | - Brittney DeWolf
- Children's National Health System, Washington, DC, United States
| | - Kristy Rose
- Discipline of Physiotherapy, Faculty of Medicine and Health, University of Sydney and the Sydney Children's Hospitals Network, Sydney, Australia
| | - Catherine Siener
- Washington University School of Medicine, St. Louis, MO, United States
| | | | | | | | - Scott Holsten
- Carolinas HealthCare System, Charlotte, United States
| | | | | | | | - Lindsay N Alfano
- Nationwide Children's Hospital, Abigail Wexner Research Inst., Columbus, OH, United States
| | - Linda Pax Lowes
- Nationwide Children's Hospital, Abigail Wexner Research Inst., Columbus, OH, United States
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon, Tyne, United Kingdom
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon, Tyne, United Kingdom.
| |
Collapse
|
7
|
Wang D, Liu XY, He QF, Zheng FZ, Chen L, Zheng Y, Zeng MH, Lin YH, Lin X, Chen HZ, Lin MT, Wang N, Wang ZQ, Lin F. Comprehensive Proteomic Analysis of Dysferlinopathy Unveiling Molecular Mechanisms and Biomarkers Linked to Pathological Progression. CNS Neurosci Ther 2024; 30:e70065. [PMID: 39350328 PMCID: PMC11442333 DOI: 10.1111/cns.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
AIMS Previous proteomics studies in dysferlinopathy muscle have been limited in scope, often utilizing 2D-electrophoresis and yielding only a small number of differential expression calls. To address this gap, this study aimed to employ high-resolution proteomics to explore the proteomic landscapes of dysferlinopathy and analyze the correlation between muscle pathological changes and alterations in protein expression in muscle biopsies. METHODS We conducted a comprehensive approach to investigate the proteomic profile and disease-associated changes in the muscle tissue proteome from 15 patients with dysferlinopathy, exhibiting varying degrees of dystrophic pathology, alongside age-matched controls. Our methodology encompasses tandem mass tag (TMT)-labeled liquid chromatography-mass spectrometry (LC-MS/MS)-based proteomics, protein-protein interaction (PPI) network analysis, weighted gene co-expression network analysis, and differential expression analysis. Subsequently, we examined the correlation between the expression of key proteins and the clinical characteristics of the patients to identify pathogenic targets associated with DYSF mutations in dysferlinopathy. RESULTS A total of 1600 differentially expressed proteins were identified, with 1321 showing high expression levels and 279 expressed at lower levels. Our investigation yields a molecular profile delineating the altered protein networks in dysferlinopathy-afflicted skeletal muscle, uncovering dysregulation across numerous cellular pathways and molecular processes, including mRNA metabolic processes, regulated exocytosis, immune response, muscle system processes, energy metabolic processes, and calcium transmembrane transport. Moreover, we observe significant associations between the protein expression of ANXA1, ANXA2, ANXA4, ANXA5, LMNA, PYGM, and the extent of histopathologic changes in muscle biopsies from patients with dysferlinopathy, validated through immunoblotting and immunofluorescence assays. CONCLUSIONS Through the aggregation of expression data from dysferlinopathy-impacted muscles exhibiting a range of pathological alterations, we identified multiple key proteins associated with the dystrophic pathology of patients with dysferlinopathy. These findings provide novel insights into the pathogenesis of dysferlinopathy and propose promising targets for future therapeutic endeavors.
Collapse
Affiliation(s)
- Di Wang
- Department of Molecular Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Center for Bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Yi Liu
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qi-Fang He
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Fu-Ze Zheng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Long Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Ying Zheng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Ming-Hui Zeng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yu-Hua Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Xin Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hai-Zhu Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Min-Ting Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ning Wang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhi-Qiang Wang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Feng Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
8
|
Lloyd EM, Crew RC, Haynes VR, White RB, Mark PJ, Jackaman C, Papadimitriou JM, Pinniger GJ, Murphy RM, Watt MJ, Grounds MD. Pilot investigations into the mechanistic basis for adverse effects of glucocorticoids in dysferlinopathy. Skelet Muscle 2024; 14:19. [PMID: 39123261 PMCID: PMC11312411 DOI: 10.1186/s13395-024-00350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by gene mutations resulting in deficiency of the membrane-associated protein dysferlin. They manifest post-growth and are characterised by muscle wasting (primarily in the limb and limb-gridle muscles), inflammation, and replacement of myofibres with adipose tissue. The precise pathomechanism for dysferlinopathy is currently unclear; as such there are no treatments currently available. Glucocorticoids (GCs) are widely used to reduce inflammation and treat muscular dystrophies, but when administered to patients with dysferlinopathy, they have unexpected adverse effects, with accelerated loss of muscle strength. METHODS To investigate the mechanistic basis for the adverse effects of GCs in dysferlinopathy, the potent GC dexamethasone (Dex) was administered for 4-5 weeks (0.5-0.75 µg/mL in drinking water) to dysferlin-deficient BLA/J and normal wild-type (WT) male mice, sampled at 5 (Study 1) or 10 months (Study 2) of age. A wide range of analyses were conducted. Metabolism- and immune-related gene expression was assessed in psoas muscles at both ages and in quadriceps at 10 months of age. For the 10-month-old mice, quadriceps and psoas muscle histology was assessed. Additionally, we investigated the impact of Dex on the predominantly slow and fast-twitch soleus and extensor digitorum longus (EDL) muscles (respectively) in terms of contractile function, myofibre-type composition, and levels of proteins related to contractile function and metabolism, plus glycogen. RESULTS At both ages, many complement-related genes were highly expressed in BLA/J muscles, and WT mice were generally more responsive to Dex than BLA/J. The effects of Dex on BLA/J mice included (i) increased expression of inflammasome-related genes in muscles (at 5 months) and (ii) exacerbated histopathology of quadriceps and psoas muscles at 10 months. A novel observation was pronounced staining for glycogen in many myofibres of the damaged quadriceps muscles, with large pale vacuolated myofibres, suggesting possible myofibre death by oncosis. CONCLUSION These pilot studies provide a new focus for further investigation into the adverse effects of GCs on dysferlinopathic muscles.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - Rachael C Crew
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| | - Vanessa R Haynes
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Robert B White
- MD Education Unit, UWA Medical School, The University of Western Australia, Perth, WA, Australia
| | - Peter J Mark
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - John M Papadimitriou
- Department of Pathology and Laboratory Medicine, UWA Medical School, The University of Western Australia, Perth, WA, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
9
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [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: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
10
|
Flores-Opazo M, Kopinke D, Helmbacher F, Fernández-Verdejo R, Tuñón-Suárez M, Lynch GS, Contreras O. Fibro-adipogenic progenitors in physiological adipogenesis and intermuscular adipose tissue remodeling. Mol Aspects Med 2024; 97:101277. [PMID: 38788527 PMCID: PMC11692456 DOI: 10.1016/j.mam.2024.101277] [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: 02/01/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Excessive accumulation of intermuscular adipose tissue (IMAT) is a common pathological feature in various metabolic and health conditions and can cause muscle atrophy, reduced function, inflammation, insulin resistance, cardiovascular issues, and unhealthy aging. Although IMAT results from fat accumulation in muscle, the mechanisms underlying its onset, development, cellular components, and functions remain unclear. IMAT levels are influenced by several factors, such as changes in the tissue environment, muscle type and origin, extent and duration of trauma, and persistent activation of fibro-adipogenic progenitors (FAPs). FAPs are a diverse and transcriptionally heterogeneous population of stromal cells essential for tissue maintenance, neuromuscular stability, and tissue regeneration. However, in cases of chronic inflammation and pathological conditions, FAPs expand and differentiate into adipocytes, resulting in the development of abnormal and ectopic IMAT. This review discusses the role of FAPs in adipogenesis and how they remodel IMAT. It highlights evidence supporting FAPs and FAP-derived adipocytes as constituents of IMAT, emphasizing their significance in adipose tissue maintenance and development, as well as their involvement in metabolic disorders, chronic pathologies and diseases. We also investigated the intricate molecular pathways and cell interactions governing FAP behavior, adipogenesis, and IMAT accumulation in chronic diseases and muscle deconditioning. Finally, we hypothesize that impaired cellular metabolic flexibility in dysfunctional muscles impacts FAPs, leading to IMAT. A deeper understanding of the biology of IMAT accumulation and the mechanisms regulating FAP behavior and fate are essential for the development of new therapeutic strategies for several debilitating conditions.
Collapse
Affiliation(s)
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, 32610, FL, USA; Myology Institute, University of Florida College of Medicine, Gainesville, FL, USA.
| | | | - Rodrigo Fernández-Verdejo
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA; Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Mauro Tuñón-Suárez
- Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Parkville 3010, Australia.
| | - Osvaldo Contreras
- Developmental and Regenerative Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia; School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia.
| |
Collapse
|
11
|
Agarwal AK, Tunison K, Horton JD, Garg A. Regulated regeneration of adipose tissue in lipodystrophic Agpat2-null mice partially ameliorates hepatic steatosis. iScience 2024; 27:109517. [PMID: 38623324 PMCID: PMC11016861 DOI: 10.1016/j.isci.2024.109517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 01/22/2024] [Accepted: 03/14/2024] [Indexed: 04/17/2024] Open
Abstract
Both humans and mice with congenital generalized lipodystrophy due to AGPAT2 deficiency develop diabetes mellitus, insulin resistance, and hepatic steatosis, which have been attributed to the near total loss of adipose tissue (AT). Here, we show that regulated AT regeneration in doxycycline (dox)-fed Tg-AT-hAGPAT2;mAgpat2-/- mice partially ameliorates hepatic steatosis at 12 weeks of age and causes reduced expression of genes involved in hepatic de novo lipogenesis despite partial (∼30-50%) AT regeneration compared to that in wild-type mice. Compared to chow-fed Tg-AT-hAGPAT2;mAgpat2-/- mice, those fed dox diet had markedly reduced serum insulin levels, suggesting an improvement in insulin resistance. Interestingly, the fasting plasma glucose levels in dox-fed Tg-AT-hAGPAT2;mAgpat2-/- mice were no different than those in chow-fed wild-type mice. Indirect calorimetry revealed normalization in the energy balance of dox-fed Tg-AT-hAGPAT2;mAgpat2-/- mice compared to that in chow-fed mice. This study's findings suggest that partial AT regeneration in lipodystrophic mice can ameliorate metabolic derangements.
Collapse
Affiliation(s)
- Anil K. Agarwal
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katie Tunison
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jay D. Horton
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Abhimanyu Garg
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
12
|
Agarwal AK, Tunison K, Vale G, McDonald JG, Li X, Horton JD, Garg A. Adipose-specific overexpression of human AGPAT2 in mice causes increased adiposity and mild hepatic dysfunction. iScience 2024; 27:108653. [PMID: 38274405 PMCID: PMC10809107 DOI: 10.1016/j.isci.2023.108653] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/11/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024] Open
Abstract
AGPAT2, a critical enzyme involved in the biosynthesis of phospholipids and triacylglycerol (TAG), is highly expressed in adipose tissue (AT). Whether overexpression of AGPAT2 in AT will result in increased TAG synthesis (obesity) and its metabolic complications remains unknown. We overexpressed human AGPAT2 specifically in AT using the adiponectin promoter and report increased mass of subcutaneous, gonadal, and brown AT in wild-type mice. Unexpectedly, overexpression of hAGPAT2 did not change the pattern of phospholipid or TAG concentration of the AT depots. Although there is an increase in liver weight, plasma aspartate aminotransferase, and plasma insulin at various time points of the study, it did not result in significant liver dysfunction. Despite increased adiposity in the Tg-AT-hAGPAT2;mAgpat2+/+ mice, there was no significant increase in TAG concentration of AT. Therefore, this study suggests a role of AGPAT2 in the generation of AT, but not for adipocyte TAG synthesis.
Collapse
Affiliation(s)
- Anil K. Agarwal
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katie Tunison
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Goncalo Vale
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey G. McDonald
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xilong Li
- Peter O’Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jay D. Horton
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Abhimanyu Garg
- Section of Nutrition and Metabolic Diseases, Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
13
|
Rufibach L, Berger K, Chakravorty S, Emmons S, Long L, Gibson G, Hegde M. Utilization of Targeted RNA-Seq for the Resolution of Variant Pathogenicity and Enhancement of Diagnostic Yield in Dysferlinopathy. J Pers Med 2023; 13:jpm13030520. [PMID: 36983702 PMCID: PMC10056012 DOI: 10.3390/jpm13030520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
For inherited diseases, obtaining a definitive diagnosis is critical for proper disease management, family planning, and participation in clinical trials. This can be challenging for dysferlinopathy due to the significant clinical overlap between the 30+ subtypes of limb–girdle muscular dystrophy (LGMD) and the large number of variants of unknown significance (VUSs) that are identified in the dysferlin gene, DYSF. We performed targeted RNA-Seq using a custom gene-panel in 77 individuals with a clinical/genetic suspicion of dysferlinopathy and evaluated all 111 identified DYSF variants according to the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) guidelines. This evaluation identified 11 novel DYSF variants and allowed for the classification of 87 DYSF variants as pathogenic/likely pathogenic, 8 likely benign, while 16 variants remained VUSs. By the end of the study, 60 of the 77 cases had a definitive diagnosis of dysferlinopathy, which was a 47% increase in diagnostic yield over the rate at study onset. This data shows the ability of RNA-Seq to assist in variant pathogenicity classification and diagnosis of dysferlinopathy and is, therefore, a type of analysis that should be considered when DNA-based genetic analysis is not sufficient to provide a definitive diagnosis.
Collapse
Affiliation(s)
- Laura Rufibach
- Jain Foundation, Inc., Seattle, WA 98115, USA; (S.E.); (L.L.)
- Correspondence:
| | - Kiera Berger
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA 30332, USA; (K.B.); (G.G.)
| | - Samya Chakravorty
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.C.); (M.H.)
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sarah Emmons
- Jain Foundation, Inc., Seattle, WA 98115, USA; (S.E.); (L.L.)
| | - Laurie Long
- Jain Foundation, Inc., Seattle, WA 98115, USA; (S.E.); (L.L.)
| | - Greg Gibson
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA 30332, USA; (K.B.); (G.G.)
| | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.C.); (M.H.)
- PerkinElmer Genomics, Global Laboratory Services, Waltham, MA 02451, USA
| |
Collapse
|
14
|
Yasa J, Reed CE, Bournazos AM, Evesson FJ, Pang I, Graham ME, Wark JR, Nijagal B, Kwan KH, Kwiatkowski T, Jung R, Weisleder N, Cooper ST, Lemckert FA. Minimal expression of dysferlin prevents development of dysferlinopathy in dysferlin exon 40a knockout mice. Acta Neuropathol Commun 2023; 11:15. [PMID: 36653852 PMCID: PMC9847081 DOI: 10.1186/s40478-022-01473-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/03/2022] [Indexed: 01/19/2023] Open
Abstract
Dysferlin is a Ca2+-activated lipid binding protein implicated in muscle membrane repair. Recessive variants in DYSF result in dysferlinopathy, a progressive muscular dystrophy. We showed previously that calpain cleavage within a motif encoded by alternatively spliced exon 40a releases a 72 kDa C-terminal minidysferlin recruited to injured sarcolemma. Herein we use CRISPR/Cas9 gene editing to knock out murine Dysf exon 40a, to specifically assess its role in membrane repair and development of dysferlinopathy. We created three Dysf exon 40a knockout (40aKO) mouse lines that each express different levels of dysferlin protein ranging from ~ 90%, ~ 50% and ~ 10-20% levels of wild-type. Histopathological analysis of skeletal muscles from all 12-month-old 40aKO lines showed virtual absence of dystrophic features and normal membrane repair capacity for all three 40aKO lines, as compared with dysferlin-null BLAJ mice. Further, lipidomic and proteomic analyses on 18wk old quadriceps show all three 40aKO lines are spared the profound lipidomic/proteomic imbalance that characterises dysferlin-deficient BLAJ muscles. Collective results indicate that membrane repair does not depend upon calpain cleavage within exon 40a and that ~ 10-20% of WT dysferlin protein expression is sufficient to maintain the muscle lipidome, proteome and membrane repair capacity to crucially prevent development of dysferlinopathy.
Collapse
Affiliation(s)
- Joe Yasa
- grid.413973.b0000 0000 9690 854XKids Neuroscience Centre, The Children’s Hospital at Westmead, Cnr Hawkesbury Road, Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ,grid.414235.50000 0004 0619 2154Functional Neuromics, Children’s Medical Research Institute, Westmead, Sydney, NSW Australia
| | - Claudia E. Reed
- grid.413973.b0000 0000 9690 854XKids Neuroscience Centre, The Children’s Hospital at Westmead, Cnr Hawkesbury Road, Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ,grid.1013.30000 0004 1936 834XDiscipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, NSW Australia
| | - Adam M. Bournazos
- grid.413973.b0000 0000 9690 854XKids Neuroscience Centre, The Children’s Hospital at Westmead, Cnr Hawkesbury Road, Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ,grid.1013.30000 0004 1936 834XDiscipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, NSW Australia
| | - Frances J. Evesson
- grid.413973.b0000 0000 9690 854XKids Neuroscience Centre, The Children’s Hospital at Westmead, Cnr Hawkesbury Road, Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ,grid.414235.50000 0004 0619 2154Functional Neuromics, Children’s Medical Research Institute, Westmead, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XDiscipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, NSW Australia
| | - Ignatius Pang
- grid.414235.50000 0004 0619 2154Synapse Proteomics, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW Australia
| | - Mark E. Graham
- grid.414235.50000 0004 0619 2154Synapse Proteomics, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW Australia
| | - Jesse R. Wark
- grid.1013.30000 0004 1936 834XOperations, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW Australia
| | - Brunda Nijagal
- grid.1008.90000 0001 2179 088XMetabolomics Australia, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - Kim H. Kwan
- grid.1008.90000 0001 2179 088XMetabolomics Australia, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - Thomas Kwiatkowski
- grid.268132.c0000 0001 0701 2416West Chester University, West Chester, PA 19383 USA
| | - Rachel Jung
- grid.412332.50000 0001 1545 0811Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210-1252 USA
| | - Noah Weisleder
- grid.412332.50000 0001 1545 0811Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210-1252 USA
| | - Sandra T. Cooper
- grid.413973.b0000 0000 9690 854XKids Neuroscience Centre, The Children’s Hospital at Westmead, Cnr Hawkesbury Road, Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ,grid.414235.50000 0004 0619 2154Functional Neuromics, Children’s Medical Research Institute, Westmead, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XDiscipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, NSW Australia
| | - Frances A. Lemckert
- grid.413973.b0000 0000 9690 854XKids Neuroscience Centre, The Children’s Hospital at Westmead, Cnr Hawkesbury Road, Hainsworth Street, Westmead, Sydney, NSW 2145 Australia ,grid.414235.50000 0004 0619 2154Functional Neuromics, Children’s Medical Research Institute, Westmead, Sydney, NSW Australia ,grid.1013.30000 0004 1936 834XDiscipline of Child and Adolescent Health, Faculty of Medicine, University of Sydney, Sydney, NSW Australia
| |
Collapse
|
15
|
Goodpaster BH, Bergman BC, Brennan AM, Sparks LM. Intermuscular adipose tissue in metabolic disease. Nat Rev Endocrinol 2022; 19:285-298. [PMID: 36564490 DOI: 10.1038/s41574-022-00784-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Intermuscular adipose tissue (IMAT) is a distinct adipose depot described in early reports as a 'fatty replacement' or 'muscle fat infiltration' that was linked to ageing and neuromuscular disease. Later studies quantifying IMAT with modern in vivo imaging methods (computed tomography and magnetic resonance imaging) revealed that IMAT is proportionately higher in men and women with type 2 diabetes mellitus and the metabolic syndrome than in people without these conditions and is associated with insulin resistance and poor physical function with ageing. In parallel, agricultural research has provided extensive insight into the role of IMAT and other muscle lipids in muscle (that is, meat) quality. In addition, studies using rodent models have shown that IMAT is a bona fide white adipose tissue depot capable of robust triglyceride storage and turnover. Insight into the importance of IMAT in human biology has been limited by the dearth of studies on its biological properties, that is, the quality of IMAT. However, in the past few years, investigations have begun to determine that IMAT has molecular and metabolic features that distinguish it from other adipose tissue depots. These studies will be critical to further decipher the role of IMAT in health and disease and to better understand its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Bryan C Bergman
- Division of Endocrinology, Diabetes, and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrea M Brennan
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
16
|
Lloyd EM, Pinniger GJ, Grounds MD, Murphy RM. Dysferlin Deficiency Results in Myofiber-Type Specific Differences in Abundances of Calcium-Handling and Glycogen Metabolism Proteins. Int J Mol Sci 2022; 24:ijms24010076. [PMID: 36613515 PMCID: PMC9820290 DOI: 10.3390/ijms24010076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by a genetic deficiency of the membrane-associated protein dysferlin, which usually manifest post-growth in young adults. The disease is characterized by progressive skeletal muscle wasting in the limb-girdle and limbs, inflammation, accumulation of lipid droplets in slow-twitch myofibers and, in later stages, replacement of muscles by adipose tissue. Previously we reported myofiber-type specific differences in muscle contractile function of 10-month-old dysferlin-deficient BLAJ mice that could not be fully accounted for by altered myofiber-type composition. In order to further investigate these findings, we examined the impact of dysferlin deficiency on the abundance of calcium (Ca2+) handling and glucose/glycogen metabolism-related proteins in predominantly slow-twitch, oxidative soleus and fast-twitch, glycolytic extensor digitorum longus (EDL) muscles of 10-month-old wild-type (WT) C57BL/6J and dysferlin-deficient BLAJ male mice. Additionally, we compared the Ca2+ activation properties of isolated slow- and fast-twitch myofibers from 3-month-old WT and BLAJ male mice. Differences were observed for some Ca2+ handling and glucose/glycogen metabolism-related protein levels between BLAJ soleus and EDL muscles (compared with WT) that may contribute to the previously reported differences in function in these BLAJ muscles. Dysferlin deficiency did not impact glycogen content of whole muscles nor Ca2+ activation of the myofilaments, although soleus muscle from 10-month-old BLAJ mice had more glycogen than EDL muscles. These results demonstrate a further impact of dysferlin deficiency on proteins associated with excitation-contraction coupling and glycogen metabolism in skeletal muscles, potentially contributing to altered contractile function in dysferlinopathy.
Collapse
Affiliation(s)
- Erin M. Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Gavin J. Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Miranda D. Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA 6009, Australia
- Correspondence:
| | - Robyn M. Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
17
|
Chernova ON, Chekmareva IA, Mavlikeev MO, Yakovlev IA, Kiyasov AP, Deev RV. Structural and ultrastructural changes in the skeletal muscles of dysferlin-deficient mice during postnatal ontogenesis. Ultrastruct Pathol 2022; 46:359-367. [PMID: 35880824 DOI: 10.1080/01913123.2022.2105464] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
A number of sarcolemma proteins are responsible for muscle fiber repair. Dysferlin encoded by the DYSF gene is one of these proteins. Dysferlin promotes membrane repair in striated muscle fibers (MFs). Mutations in DYSF lead to loss of or decreased dysferlin expression, impaired membrane repair in MF, and its destruction, clinically manifesting as dysferlinopathy. Preclinical studies of cell and gene therapies aimed at restoring impaired muscle regeneration require well-characterized small animal models. Our investigation aimed to distinguish the histopathological features of a mouse strain lacking dysferlin expression (Bla/J strain). Ultrastructural changes in the sarcolemma, mitochondria and contractile apparatus were observed. It was shown that postnatal histogenesis of skeletal muscles in genetically determined dysferlin deficiency is characterized by a higher proportion of necrotic muscle fibers, compensatory hypertrophy of muscle fibers with their subsequent atrophy, and decreases in proliferative activity and the level of myogenic differentiation of myogenic progenitor cells compared to wild-type mice (C57Bl/6).
Collapse
Affiliation(s)
- O N Chernova
- Human Morphology Department, North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, Russian Federation.,Pathology and Forensic Medicine Department, Saint-Petersburg Medico-Social Institute, Saint-Petersburg, Russian Federation
| | - I A Chekmareva
- A.V. Vishnevsky National Medical Research Center of Surgery, Moscow, Russian Federation
| | - M O Mavlikeev
- Pathology Department, North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, Russian Federation
| | - I A Yakovlev
- Genotarget LLC, Moscow, Russian Federation.,Human Stem Cell Institute PJSC, Moscow, Russian Federation
| | - A P Kiyasov
- Morphology and General Pathology Department, Kazan (Volga region) Federal University, Kazan, Russian Federation
| | - R V Deev
- Pathology Department, North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, Russian Federation.,Human Stem Cell Institute PJSC, Moscow, Russian Federation
| |
Collapse
|
18
|
Donen GS, White Z, Sauge E, Ritso M, Theret M, Boyd J, Devlin AM, Rossi FMV, Bernatchez P. Thermoneutral Housing and a Western Diet Combination Exacerbates Dysferlin-Deficient Muscular Dystrophy. Muscle Nerve 2022; 66:513-522. [PMID: 35859452 DOI: 10.1002/mus.27680] [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: 12/20/2021] [Revised: 07/10/2022] [Accepted: 07/17/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION/AIMS Most mouse models of muscular dystrophy (MD) show mild phenotypes, which limits the translatability of experimental therapies to patients. A growing body of evidence suggests that MD is accompanied by metabolic abnormalities that could potentially exacerbate the primary muscle wasting process. Since thermoneutral (TN) housing of mice (~30°C) has been shown to affect many metabolic parameters, particularly when combined with a Western diet (WD), our aim was to determine whether the combination of TN and WD exacerbates muscle wasting in dysferlin-deficient BLAJ mice, a common model of limb-girdle MD type 2b (LGMD2b). METHODS Two-month-old wild-type (WT) and BLAJ mice were housed at TN or room temperature (RT) and fed a WD or regular chow for 9 months. Ambulatory function, muscle histology, and protein immunoblots of skeletal muscle were assessed. RESULTS BLAJ mice at RT and fed a chow diet showed normal ambulation function similar to WT mice, whereas 90 % of BLAJ mice under WD and TN combination showed ambulatory dysfunction (P<0.001), and an up to 4.1-fold increase in quadriceps and gastrocnemius fat infiltration. Western blotting revealed decreased autophagy marker microtubules-associated protein 1 light chain 3-B (LC3BII/LC3BI) ratio and up-regulation of AKT and ribosomal protein S6 (rpS6) phosphorylation, suggesting inefficient cellular debris and protein clearance in TN BLAJ mice fed a WD. Male and female BLAJ mice under TN and WD combination showed heterogenous fibro-fatty infiltrate composition. DISCUSSION TN and WD combination exacerbates rodent LGMD2b without affecting WT mice. This improves rodent modeling of human MD and helps elucidate how metabolic abnormalities may play a causal role in muscle wasting.
Collapse
Affiliation(s)
- Graham S Donen
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Zoe White
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Elodie Sauge
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Morten Ritso
- The Biomedical Research Centre, UBC, 2222 Health Sciences Mall, Vancouver, Canada
| | - Marine Theret
- The Biomedical Research Centre, UBC, 2222 Health Sciences Mall, Vancouver, Canada
| | - John Boyd
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Angela M Devlin
- University of British Columbia (UBC) Department of Pediatrics, BC Children's Hospital Research Institute, Vancouver, Canada
| | - Fabio M V Rossi
- The Biomedical Research Centre, UBC, 2222 Health Sciences Mall, Vancouver, Canada
| | - Pascal Bernatchez
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| |
Collapse
|
19
|
White Z, Theret M, Milad N, Tung LW, Chen WWH, Sirois MG, Rossi F, Bernatchez P. Cholesterol absorption blocker ezetimibe prevents muscle wasting in severe dysferlin-deficient and mdx mice. J Cachexia Sarcopenia Muscle 2022; 13:544-560. [PMID: 34927367 PMCID: PMC8818667 DOI: 10.1002/jcsm.12879] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Muscular dystrophy (MD) causes muscle wasting and is often lethal in patients due to a lack of proven therapies. In contrast, mouse models of MD are notoriously mild. We have previously shown severe human-like muscle pathology in mdx [Duchenne MD (DMD)] and dysferlin-deficient limb-girdle MD type 2B (LGMD2B) mice by inactivating the gene encoding for apolipoprotein E (ApoE), a lipid transporter synthesized by the liver, brain and adipocytes to regulate lipid and fat metabolism. Having recently established that human DMD is a novel type of primary genetic dyslipidaemia with elevated cholesterol, we sought to determine whether cholesterol could exacerbate the muscle wasting process observed in severe rodent MD. METHODS Severe mdx and dysferlin knock-out mice lacking ApoE were treated with ezetimibe (15 mg/kg/day), a clinically approved drug exhibiting few pleiotropic effects. In separate studies, dietary cholesterol was raised (from 0.2% to 2% cholesterol) in combination with experimental micro-injury and direct cholesterol injection assays. Muscles were assessed histologically for changes in collagen and adipocyte infiltration and both transcriptomic and cellular changes by RNA-seq and fluorescence-activated cell sorting analysis. RESULTS Treatment of severe DMD and LGMD2B mice with ezetimibe completely prevented clinical signs of ambulatory dysfunction (0% incidence vs. 33% for vehicle treatment; P < 0.05). Histological analyses revealed that ezetimibe-reduced fibro-fatty infiltration up to 84% and 63% in severely affected triceps (P ≤ 0.0001) and gastrocnemius (P ≤ 0.003) muscles, resulting in a respective 1.9-fold and 2.2-fold retention of healthy myofibre area (P ≤ 0.0001). Additionally, raising dietary cholesterol and thus concentrations of plasma low-density lipoprotein-associated cholesterol (by 250%; P < 0.0001) reduced overall survivability (by 100%; P < 0.001) and worsened muscle damage in the LGMD2B triceps by 767% (P < 0.03). Micro-pin-induced mechanical injury in LGMD2B mice fed a high cholesterol diet exacerbated muscle damage by 425% (P < 0.03) and increased macrophage recruitment (by 98%; P = 0.03) compared with those injured on a chow diet. Parallel RNA-seq analyses revealed that injury in cholesterol-fed mice also modulated the expression of 3671 transcripts (1953 up-regulated), with fibrogenic, inflammatory and programmed cell death-associated pathways among the most enriched. Mice lacking dysferlin also displayed heightened muscle necrosis (by 123%; P < 0.0001) following a direct intramuscular injection of cholesterol compared with control mice. CONCLUSIONS Cholesterol exacerbates rodent MD. Specific inhibition of cholesterol absorption with ezetimibe may safely attenuate human MD severity and delay death.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - William Wei-Han Chen
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Martin G Sirois
- Montreal Heart Institute, Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Fabio Rossi
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW The goal of this review is to highlight the need for new biomarkers for the diagnosis and treatment of musculoskeletal disorders, especially osteoporosis and sarcopenia. These conditions are characterized by loss of bone and muscle mass, respectively, leading to functional deterioration and the development of disabilities. Advances in high-resolution lipidomics platforms are being used to help identify new lipid biomarkers for these diseases. RECENT FINDINGS It is now well established that bone and muscle have important endocrine functions, including the release of bioactive factors in response to mechanical and biochemical stimuli. Bioactive lipids are a prominent set of these factors and some of these lipids are directly related to the mass and function of bone and muscle. Recent lipidomics studies have shown significant dysregulation of lipids in aged muscle and bone, including alterations in diacylglycerols and ceramides. Studies have shown that alterations in some types of plasma lipids are associated with aging including reduced bone mineral density and the occurrence of osteoporosis. Musculoskeletal disorders are a major burden in our society, especially for older adults. The development and application of new lipidomics methods is making significant advances in identifying new biomarkers for these diseases. These studies will not only lead to improved detection, but new mechanistic insights that could lead to new therapeutic targets and interventions.
Collapse
Affiliation(s)
- Chenglin Mo
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, USA.
| | - Yating Du
- Bone-Muscle Research Center, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, USA
| | - Thomas M O'Connell
- Department of Otolaryngology, Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
21
|
Blockade of Hemichannels Normalizes the Differentiation Fate of Myoblasts and Features of Skeletal Muscles from Dysferlin-Deficient Mice. Int J Mol Sci 2020; 21:ijms21176025. [PMID: 32825681 PMCID: PMC7503700 DOI: 10.3390/ijms21176025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 01/18/2023] Open
Abstract
Dysferlinopathies are muscle dystrophies caused by mutations in the gene encoding dysferlin, a relevant protein for membrane repair and trafficking. These diseases are untreatable, possibly due to the poor knowledge of relevant molecular targets. Previously, we have shown that human myofibers from patient biopsies as well as myotubes derived from immortalized human myoblasts carrying a mutated form of dysferlin express connexin proteins, but their relevance in myoblasts fate and function remained unknown. In the present work, we found that numerous myoblasts bearing a mutated dysferlin when induced to acquire myogenic commitment express PPARγ, revealing adipogenic instead of myogenic commitment. These cell cultures presented many mononucleated cells with fat accumulation and within 48 h of differentiation formed fewer multinucleated cells. In contrast, dysferlin deficient myoblasts treated with boldine, a connexin hemichannels blocker, neither expressed PPARγ, nor accumulated fat and formed similar amount of multinucleated cells as wild type precursor cells. We recently demonstrated that myofibers of skeletal muscles from blAJ mice (an animal model of dysferlinopathies) express three connexins (Cx39, Cx43, and Cx45) that form functional hemichannels (HCs) in the sarcolemma. In symptomatic blAJ mice, we now show that eight-week treatment with a daily dose of boldine showed a progressive recovery of motor activity reaching normality. At the end of this treatment, skeletal muscles were comparable to those of wild type mice and presented normal CK activity in serum. Myofibers of boldine-treated blAJ mice also showed strong dysferlin-like immunoreactivity. These findings reveal that muscle dysfunction results from a pathophysiologic mechanism triggered by mutated dysferlin and downstream connexin hemichannels expressed de novo lead to a drastic reduction of myogenesis and favor muscle damage. Thus, boldine could represent a therapeutic opportunity to treat dysfernilopathies.
Collapse
|
22
|
Zhang L, Li F, Guo Q, Duan Y, Wang W, Zhong Y, Yang Y, Yin Y. Leucine Supplementation: A Novel Strategy for Modulating Lipid Metabolism and Energy Homeostasis. Nutrients 2020; 12:E1299. [PMID: 32370170 PMCID: PMC7282259 DOI: 10.3390/nu12051299] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Lipid metabolism is an important and complex biochemical process involved in the storage of energy and maintenance of normal biological functions. Leucine, a branched amino acid, has anti-obesity effects on glucose tolerance, lipid metabolism, and insulin sensitivity. Leucine also modulates mitochondrial dysfunction, representing a new strategy to target aging, neurodegenerative disease, obesity, diabetes, and cardiovascular disease. Although various studies have been carried out, much uncertainty still exists and further studies are required to fully elucidate the relationship between leucine and lipid metabolism. This review offers an up-to-date report on leucine, as key roles in both lipid metabolism and energy homeostasis in vivo and in vitro by acceleration of fatty acid oxidation, lipolysis, activation of the adenosine 5'-monophosphate-activated protein kinase (AMPK)-silent information regulator of transcription 1 (SIRT1)-proliferator-activated receptor γ coactivator-1α (PGC-1α) pathway, synthesis, and/or secretion of adipokines and stability of the gut microbiota.
Collapse
Affiliation(s)
- Lingyu Zhang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
| | - Wenlong Wang
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
- Laboratory of Animal Nutrition and Human Health, School of Biology, Hunan Normal University, Changsha 410018, China
| | - Yinzhao Zhong
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou 510642, China;
| | - Yuhuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China;
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (L.Z.); (Q.G.); (Y.D.); (W.W.); (Y.Y.)
| |
Collapse
|