1
|
Hamzeh A, Saelee L, Yongsawatdigul J. Physico-chemical properties of natural actomyosin from breast and thigh meat of fast- and slow-growing chicken: a comparative study. Poult Sci 2024; 103:104153. [PMID: 39153267 PMCID: PMC11471090 DOI: 10.1016/j.psj.2024.104153] [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: 03/16/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/19/2024] Open
Abstract
Physico-chemical behaviors of natural actomyosin (NAM) from slow-growing Korat chicken (KC) breast and thigh at low (0.2 M) and high (0.6 M) NaCl concentrations were evaluated and compared to those from their corresponding muscles in fast-growing broiler chicken (BC). NAM from KC breast and thigh meat showed higher solubility than their corresponding in BC (p < 0.05). Breast NAM from both chickens showed the highest solubility at 0.4 M NaCl, while thigh NAMs showed the highest solubility at 0.8 M (p < 0.05). SDS-PAGE revealed higher protein extractability for breast NAMs at low ionic strength, regardless of breed and structural protein, particularly troponin, appeared to vary within muscle and breed. NAM from KC showed higher Ca2+-ATPase activity, surface hydrophobicity, but lower total sulfhydryl groups content (p < 0.05). Particle size of NAM solutions varied with ionic strength, in which KC-NAM showed larger size than at lower ionic strength, while BC-NAM appeared to be greater at higher ionic strength. NAM from KC breast showed higher thermal stability as higher initial (T0) and maximum (Tmax) denaturation temperatures of 48.4 and 54.8°C, respectively, recorded by microdifferential scanning calorimetry. The KC-NAM, particularly from breast, exhibited lower turbidity within 40-50°C, while turbidity increased in BC samples at low ionic strength when heated at 60°C. Dynamic rheology revealed different storage modulus (G') depending on breed, muscle type and ionic strength. Breast BC-NAM formed more elastic gel with higher end point G' at 80°C (Ge'; p < 0.05). Ionic strength showed reverse effects on different breeds as a stronger Ge' value achieved in KC- NAM at low ionic strength, while high ionic strength induced stronger gel in BC samples. Aggregation of NAMs began at lower temperatures at higher ionic strength and actin dissociation probably occurred in breast NAM from KC as observed by a drop of G' at around 70°C. The results of this study revealed differences between NAM of the two breeds, by which higher gel elasticity are expected in KC at lower ionic strength.
Collapse
Affiliation(s)
- Ali Hamzeh
- School of Food Technology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Lobdaw Saelee
- School of Food Technology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Jirawat Yongsawatdigul
- School of Food Technology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
2
|
Liu Y, Shan Y, Pang L, Tang Y, Zhang M, Tu Y, Ji G, Ju X, Jiang H, Xie B, Shi S, Shu J. METTL16 inhibits differentiation and promotes proliferation and slow myofibers formation in chicken myoblasts. Poult Sci 2024; 103:104384. [PMID: 39418792 DOI: 10.1016/j.psj.2024.104384] [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: 07/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
N6-methyladenosine (m6A) plays a crucial regulatory role in muscle growth and development. In our previous studies, we identified a m6A methyltransferase, Methyltransferase like 16 (METTL16), which is associated with chicken muscle development and muscle fiber type conversion. To further understand the regulatory role of METTL16 in chicken muscle function, we analyzed its expression in muscle tissues with different myofiber type compositions and in chicken primary myoblasts (CPMs) at various stages. We also manipulated METTL16 expression in CPMs to examine its effects on cell proliferation, differentiation, muscle fiber type formation, and global m6A RNA methylation status. Our results showed that METTL16 expression increased during myoblast proliferation and gradually decreased in the late differentiation stage. Furthermore, METTL16 exhibited specific expression in slow-twitch muscles. Cell Counting Kit-8 assays, 5-Ethynyl-2'-deoxyuridine staining, RT-qPCR, Western blot, and immunofluorescence analyses showed that METTL16 promotes myoblast proliferation while inhibiting myoblast differentiation. We also observed that METTL16 induces the upregulation of slow-twitch myosin heavy chain (MyHC) and slow-twitch-specific genes in myotubes, while downregulating fast-twitch MyHC and fast-twitch-specific genes. Furthermore, both interference and overexpression of METTL16 led to changes in overall cellular m6A modification levels and Methyltransferase like 3 (METTL3) expression levels. These findings confirm that METTL16 plays a key role in myoblast proliferation, differentiation, and muscle fiber type formation in chickens. Considering the role of myoblasts in chicken muscle growth and meat quality regulation, METTL16 may serve as a key target for molecular selection in chicken meat traits.
Collapse
Affiliation(s)
- Yifan Liu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Yanju Shan
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Lichuan Pang
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Yanfei Tang
- Guangxi Fufeng Farming Group Co., Ltd., Nanning, 530024, Guangxi, China
| | - Ming Zhang
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Yunjie Tu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Gaige Ji
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Xiaojun Ju
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Hualian Jiang
- Guangxi Fufeng Farming Group Co., Ltd., Nanning, 530024, Guangxi, China
| | - Binghong Xie
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Shiying Shi
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China
| | - Jingting Shu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Jiangsu Institute of Poultry Science, Yangzhou, 225125, Jiangsu, China.
| |
Collapse
|
3
|
Dada A, Habibi J, Naz H, Chen D, Lastra G, Bostick BP, Whaley-Connell A, Hill MA, Sowers JR, Jia G. Enhanced ECCD36 signaling promotes skeletal muscle insulin resistance in female mice. Am J Physiol Endocrinol Metab 2024; 327:E533-E543. [PMID: 39196801 PMCID: PMC11482271 DOI: 10.1152/ajpendo.00246.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 08/30/2024]
Abstract
Consumption of a Western diet (WD) increases CD36 expression in vascular, hepatic, and skeletal muscle tissues promoting lipid metabolic disorders and insulin resistance. We further examined the role of endothelial cell-specific CD36 (ECCD36) signaling in contributing to skeletal muscle lipid metabolic disorders, insulin resistance, and their underlying molecular mechanisms. Female ECCD36 wild-type (ECCD36+/+) and knock-out (ECCD36-/-) mice, aged 6 wk, were provided with either a WD or a standard chow diet for a duration of 16 wk. ECCD36+/+ WD mice were characterized by elevated fasting plasma glucose and insulin levels, increased homeostatic model assessment for insulin resistance, and glucose intolerance that was blunted in ECCD36-/- mice. Improved insulin sensitivity in ECCD36-/- mice was characterized by increased phosphoinositide 3-kinases/protein kinase B signaling that further augmented glucose transporter type 4 expression and glucose uptake. Meanwhile, 16 wk of WD feeding also increased skeletal muscle free fatty acid (FFA) and lipid accumulation, without any observed changes in plasma FFA levels. These lipid metabolic disorders were blunted in ECCD36-/- mice. Moreover, ECCD36 also mediated in vitro palmitic acid-induced lipid accumulation in cultured ECs, subsequently leading to the release of FFAs into the culture media. Furthermore, consumption of a WD increased FFA oxidation, mitochondrial dysfunction, impaired mitochondrial respiratory, skeletal muscle fiber type transition, and fibrosis. These WD-induced abnormalities were blunted in ECCD36-/- mice. These findings demonstrate that endothelial-specific ECCD36 signaling participates in skeletal muscle FFA uptake, ectopic lipid accumulation, mitochondrial dysfunction, insulin resistance, and associated skeletal muscle dysfunction in diet-induced obesity.NEW & NOTEWORTHY ECCD36 exerts "extra endothelial cell" actions in skeletal muscle insulin resistance. ECCD36 is a major mediator of Western diet-induced lipid metabolic disorders and insulin resistance in skeletal muscle. Mitochondrial dysfunction is associated with diet-induced CD36 activation and related skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Austin Dada
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Javad Habibi
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Huma Naz
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Dongqing Chen
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Guido Lastra
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Brian P Bostick
- Department of Medicine-Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Adam Whaley-Connell
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
- Department of Medicine-Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - James R Sowers
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
- Department of Medicine-Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
- Department of Medicine-Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Guanghong Jia
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
4
|
Lin Y, Shi H, Yang R, Li S, Tang J, Li S. A transcriptomic analysis of incisional hernia based on high-throughput sequencing technology. Hernia 2024; 28:1899-1907. [PMID: 39073735 DOI: 10.1007/s10029-024-03116-7] [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: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Incisional hernia is a common postoperative complication; however, few transcriptomic studies have been conducted on it. In this study, we used second-generation high-throughput sequencing to explore the pathogenesis and potential therapeutic targets of incisional hernias. METHODS Superficial fasciae were collected from 15 patients without hernia and 21 patients with an incisional hernia. High-throughput sequencing of the fascia was performed to generate an expression matrix. We analyzed the matrix to identify differentially expressed genes (DEGs) and performed gene ontology and enrichment analyses of these DEGs. Additionally, an external dataset was utilized to identify key DEGs. RESULTS We identified 1,823 DEGs closely associated with extracellular matrix (ECM) imbalance, bacterial inflammatory response, and fibrillar collagen trimerization. TNNT3, CMAY5, ATP1B4, ASB5, CILP, SIX4, FBN1 and FNDC5 were identified as key DEGs at the intersection of the two expression matrices. Moreover, non-alcoholic fatty liver disease-related, TNF, and IL-17 signaling pathways were identified as key enrichment pathways. CONCLUSIONS We identified eight key DEGs and three pathways associated with incisional hernias. Our findings offer new insights into the pathogenesis of incisional hernias and highlight potential targets for their prevention and treatment.
Collapse
Affiliation(s)
- Yiming Lin
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Hekai Shi
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Rongduo Yang
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Shaochun Li
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Jianxiong Tang
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China
| | - Shaojie Li
- Department of General Surgery, Fudan University Affiliated Huadong Hosptial, No.221, West Yan'an Road, Jing'an District, Shanghai, 200040, Republic of China.
| |
Collapse
|
5
|
Joyce W, He K, Zhang M, Ogunsola S, Wu X, Joseph KT, Bogomolny D, Yu W, Springer MS, Xie J, Signore AV, Campbell KL. Genetic excision of the regulatory cardiac troponin I extension in high-heart rate mammal clades. Science 2024; 385:1466-1471. [PMID: 39325895 DOI: 10.1126/science.adi8146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 01/18/2024] [Accepted: 07/17/2024] [Indexed: 09/28/2024]
Abstract
Mammalian cardiac troponin I (cTnI) contains a highly conserved amino-terminal extension harboring protein kinase A targets [serine-23 and -24 (Ser23/24)] that are phosphorylated during β-adrenergic stimulation to defend diastolic filling by means of an increased cardiomyocyte relaxation rate. In this work, we show that the Ser23/24-encoding exon 3 of TNNI3 was pseudoexonized multiple times in shrews and moles to mimic Ser23/24 phosphorylation without adrenergic stimulation, facilitating the evolution of exceptionally high resting heart rates (~1000 beats per minute). We further reveal alternative exon 3 splicing in distantly related bat families and confirm that both cTnI splice variants are incorporated into cardiac myofibrils. Because exon 3 of human TNNI3 exhibits a relatively low splice strength score, our findings offer an evolutionarily informed strategy to excise this exon to improve diastolic function during heart failure.
Collapse
Affiliation(s)
- William Joyce
- Department of Biology - Zoophysiology, Aarhus University, 8000 Aarhus C, Denmark
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Kai He
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Mengdie Zhang
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Samuel Ogunsola
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Xini Wu
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Kelvin T Joseph
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - David Bogomolny
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Wenhua Yu
- Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong 510006, China
| | - Mark S Springer
- Department of Evolution, Ecology and Organismal Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Jiuyong Xie
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Anthony V Signore
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Kevin L Campbell
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
6
|
Vahle B, Heilmann L, Schauer A, Augstein A, Jarabo MEP, Barthel P, Mangner N, Labeit S, Bowen TS, Linke A, Adams V. Modulation of Titin and Contraction-Regulating Proteins in a Rat Model of Heart Failure with Preserved Ejection Fraction: Limb vs. Diaphragmatic Muscle. Int J Mol Sci 2024; 25:6618. [PMID: 38928324 PMCID: PMC11203682 DOI: 10.3390/ijms25126618] [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: 05/24/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by biomechanically dysfunctional cardiomyocytes. Underlying cellular changes include perturbed myocardial titin expression and titin hypophosphorylation leading to titin filament stiffening. Beside these well-studied alterations at the cardiomyocyte level, exercise intolerance is another hallmark of HFpEF caused by molecular alterations in skeletal muscle (SKM). Currently, there is a lack of data regarding titin modulation in the SKM of HFpEF. Therefore, the aim of the present study was to analyze molecular alterations in limb SKM (tibialis anterior (TA)) and in the diaphragm (Dia), as a more central SKM, with a focus on titin, titin phosphorylation, and contraction-regulating proteins. This study was performed with muscle tissue, obtained from 32-week old female ZSF-1 rats, an established a HFpEF rat model. Our results showed a hyperphosphorylation of titin in limb SKM, based on enhanced phosphorylation at the PEVK region, which is known to lead to titin filament stiffening. This hyperphosphorylation could be reversed by high-intensity interval training (HIIT). Additionally, a negative correlation occurring between the phosphorylation state of titin and the muscle force in the limb SKM was evident. For the Dia, no alterations in the phosphorylation state of titin could be detected. Supported by data of previous studies, this suggests an exercise effect of the Dia in HFpEF. Regarding the expression of contraction regulating proteins, significant differences between Dia and limb SKM could be detected, supporting muscle atrophy and dysfunction in limb SKM, but not in the Dia. Altogether, these data suggest a correlation between titin stiffening and the appearance of exercise intolerance in HFpEF, as well as a differential regulation between different SKM groups.
Collapse
Affiliation(s)
- Beatrice Vahle
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Leonard Heilmann
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Antje Schauer
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Antje Augstein
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Maria-Elisa Prieto Jarabo
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Peggy Barthel
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Norman Mangner
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Siegfried Labeit
- DZHK Partner Site Mannheim-Heidelberg, Medical Faculty Mannheim, University of Heidelberg, 68169 Mannheim, Germany;
- Myomedix GmbH, 69151 Neckargemünd, Germany
| | - T. Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK;
| | - Axel Linke
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| | - Volker Adams
- Heart Center Dresden, Laboratory of Molecular and Experimental Cardiology, TU Dresden, 01307 Dresden, Germany; (B.V.); (L.H.); (A.S.); (A.A.); (M.-E.P.J.); (P.B.); (N.M.); (A.L.)
| |
Collapse
|
7
|
Clark SR, Wilton LR, Dawson JL, Chiew K, Jawahar MC, Toben C, Pukala T, Ajaero C, Saleem M. Dotting the I's and crossing the T's: A South Australian perspective on variability in troponin thresholds for myocarditis risk in clozapine treatment. Schizophr Res 2024; 268:114-117. [PMID: 37516549 DOI: 10.1016/j.schres.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/08/2023] [Accepted: 07/09/2023] [Indexed: 07/31/2023]
Affiliation(s)
- Scott R Clark
- University of Adelaide, Discipline of Psychiatry, Adelaide, South Australia, Australia; Basil Hetzel Institute, Woodville, South Australia, Australia; Central Adelaide Local Health Network, Adelaide, South Australia, Australia.
| | - Lisa R Wilton
- Office of the Chief Psychiatrist, Adelaide, South Australia, Australia
| | - Jessica L Dawson
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; SA Pharmacy, Southern Adelaide Local Health Network, Bedford Park, South Australia, Australia
| | - Kim Chiew
- Central Adelaide Local Health Network, Adelaide, South Australia, Australia
| | - M Catharine Jawahar
- University of Adelaide, Discipline of Psychiatry, Adelaide, South Australia, Australia
| | - Catherine Toben
- University of Adelaide, Discipline of Psychiatry, Adelaide, South Australia, Australia
| | - Tara Pukala
- Department of Chemistry, School of Physical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | | | | |
Collapse
|
8
|
Johnston CI, Silva A, Hodgson W, Isbister GK. Investigating skeletal muscle biomarkers for the early detection of Australian myotoxic snake envenoming: an animal model pilot study. Clin Toxicol (Phila) 2024; 62:280-287. [PMID: 38804832 DOI: 10.1080/15563650.2024.2349690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Myotoxicity is an important toxidrome that can occur with envenoming from multiple Australian snake types. Early antivenom administration is an important strategy to reduce the incidence and severity of myotoxicity. The current gold standard biomarker, serum creatine kinase activity, does not rise early enough to facilitate early antivenom administration. Several other skeletal muscle biomarkers have shown promise in other animal models and scenarios. The aim of this study was to examine the predictive values of six skeletal muscle biomarkers in a rat model of Australian snake myotoxicity. METHODS Sprague-Dawley rats were anaesthetised and administered either Pseudechis porphyriacus (red-bellied black snake) or Notechis scutatus (tiger snake) venom, or normal saline via intramuscular injection. Blood samples were collected. Assays were performed for serum creatine kinase skeletal muscle troponin-I concentration, skeletal muscle troponin-C concentration, myoglobin activity, skeletal muscle myosin light chain-1 concentration, and creatine kinase-MM activity. Serum markers were plotted against time, with comparison of area under the concentration (or activity)-time curve. The predictive values of six skeletal muscle biomarkers were examined using receiver operating characteristic curves. RESULTS There was no difference in area under the serum creatine kinase activity-time curve between venom and control groups. Serum creatine kinase-MM activity rose early in the venom treated rats, which had a significantly greater area under the serum activity-time curve. No difference in area under the serum concentration-time curve was demonstrated for the other biomarkers. Creatine kinase-MM activity had a superior predictive values than creatine kinase activity at 0-4 hours and 0-10 hours after venom administration, as indicated by area under the receiver operating characteristic curves (95 per cent confidence intervals) of 0.91 (0.78-1.00) and 0.88 (0.73-1.00) versus 0.79 (0.63-0.95) and 0.66 (0.51-0.80). DISCUSSION The limitations of serum creatine kinase activity in early detection of myotoxicity were demonstrated in this rat model. CONCLUSION Serum creatine kinase-MM activity was superior for early detection of Australian myotoxic snake envenoming.
Collapse
Affiliation(s)
| | - Anjana Silva
- Department of Parasitology, Faculty of Medicine and Allied Sciences, Rajarata University of Sri Lanka, Anuradhapura, Sri Lanka
| | - Wayne Hodgson
- Monash Venom Group, Department of Pharmacology, Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Geoffrey K Isbister
- Clinical Toxicology Research Group, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
9
|
Nikonova E, DeCata J, Canela M, Barz C, Esser A, Bouterwek J, Roy A, Gensler H, Heß M, Straub T, Forne I, Spletter ML. Bruno 1/CELF regulates splicing and cytoskeleton dynamics to ensure correct sarcomere assembly in Drosophila flight muscles. PLoS Biol 2024; 22:e3002575. [PMID: 38683844 PMCID: PMC11081514 DOI: 10.1371/journal.pbio.3002575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 05/09/2024] [Accepted: 03/04/2024] [Indexed: 05/02/2024] Open
Abstract
Muscles undergo developmental transitions in gene expression and alternative splicing that are necessary to refine sarcomere structure and contractility. CUG-BP and ETR-3-like (CELF) family RNA-binding proteins are important regulators of RNA processing during myogenesis that are misregulated in diseases such as Myotonic Dystrophy Type I (DM1). Here, we report a conserved function for Bruno 1 (Bru1, Arrest), a CELF1/2 family homolog in Drosophila, during early muscle myogenesis. Loss of Bru1 in flight muscles results in disorganization of the actin cytoskeleton leading to aberrant myofiber compaction and defects in pre-myofibril formation. Temporally restricted rescue and RNAi knockdown demonstrate that early cytoskeletal defects interfere with subsequent steps in sarcomere growth and maturation. Early defects are distinct from a later requirement for bru1 to regulate sarcomere assembly dynamics during myofiber maturation. We identify an imbalance in growth in sarcomere length and width during later stages of development as the mechanism driving abnormal radial growth, myofibril fusion, and the formation of hollow myofibrils in bru1 mutant muscle. Molecularly, we characterize a genome-wide transition from immature to mature sarcomere gene isoform expression in flight muscle development that is blocked in bru1 mutants. We further demonstrate that temporally restricted Bru1 rescue can partially alleviate hypercontraction in late pupal and adult stages, but it cannot restore myofiber function or correct structural deficits. Our results reveal the conserved nature of CELF function in regulating cytoskeletal dynamics in muscle development and demonstrate that defective RNA processing due to misexpression of CELF proteins causes wide-reaching structural defects and progressive malfunction of affected muscles that cannot be rescued by late-stage gene replacement.
Collapse
Affiliation(s)
- Elena Nikonova
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Jenna DeCata
- School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, Missouri, United States of America
| | - Marc Canela
- Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Christiane Barz
- Muscle Dynamics Group, Max Planck Institute of Biochemistry, München, Germany
| | - Alexandra Esser
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Jessica Bouterwek
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Akanksha Roy
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Heidemarie Gensler
- Department of Systematic Zoology, Biocenter, Faculty of Biology, Ludwig-Maximilians-Universität München, München, Germany
| | - Martin Heß
- Department of Systematic Zoology, Biocenter, Faculty of Biology, Ludwig-Maximilians-Universität München, München, Germany
| | - Tobias Straub
- Biomedical Center, Bioinformatics Core Unit, Ludwig-Maximilians-Universität München, München, Germany
| | - Ignasi Forne
- Biomedical Center, Protein Analysis Unit, Ludwig-Maximilians-Universität München, München, Germany
| | - Maria L. Spletter
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, München, Germany
- School of Science and Engineering, Division of Biological and Biomedical Systems, Kansas City, Missouri, United States of America
| |
Collapse
|
10
|
Das S, Hilman MC, Yang F, Mourkioti F, Yang W, Cullen DK. Motor neurons and endothelial cells additively promote development and fusion of human iPSC-derived skeletal myocytes. Skelet Muscle 2024; 14:5. [PMID: 38454511 PMCID: PMC10921694 DOI: 10.1186/s13395-024-00336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Neurovascular cells have wide-ranging implications on skeletal muscle biology regulating myogenesis, maturation, and regeneration. Although several in vitro studies have investigated how motor neurons and endothelial cells interact with skeletal myocytes independently, there is limited knowledge about the combined effect of neural and vascular cells on muscle maturation and development. METHODS Here, we report a triculture system comprising human-induced pluripotent stem cell (iPSC)-derived skeletal myocytes, human iPSC-derived motor neurons, and primary human endothelial cells maintained under controlled media conditions. Briefly, iPSCs were differentiated to generate skeletal muscle progenitor cells (SMPCs). These SMPCs were seeded at a density of 5 × 104 cells/well in 12-well plates and allowed to differentiate for 7 days before adding iPSC-derived motor neurons at a concentration of 0.5 × 104 cells/well. The neuromuscular coculture was maintained for another 7 days in coculture media before addition of primary human umbilical vein endothelial cells (HUVEC) also at 0.5 × 104 cells/well. The triculture was maintained for another 7 days in triculture media comprising equal portions of muscle differentiation media, coculture media, and vascular media. Extensive morphological, genetic, and molecular characterization was performed to understand the combined and individual effects of neural and vascular cells on skeletal muscle maturation. RESULTS We observed that motor neurons independently promoted myofiber fusion, upregulated neuromuscular junction genes, and maintained a molecular niche supportive of muscle maturation. Endothelial cells independently did not support myofiber fusion and downregulated expression of LRP4 but did promote expression of type II specific myosin isoforms. However, neurovascular cells in combination exhibited additive increases in myofiber fusion and length, enhanced production of Agrin, along with upregulation of several key genes like MUSK, RAPSYN, DOK-7, and SLC2A4. Interestingly, more divergent effects were observed in expression of genes like MYH8, MYH1, MYH2, MYH4, and LRP4 and secretion of key molecular factors like amphiregulin and IGFBP-4. CONCLUSIONS Neurovascular cells when cultured in combination with skeletal myocytes promoted myocyte fusion with concomitant increase in expression of various neuromuscular genes. This triculture system may be used to gain a deeper understanding of the effects of the neurovascular niche on skeletal muscle biology and pathophysiology.
Collapse
Affiliation(s)
- Suradip Das
- Department of Neurosurgery, Center for Brain Injury & Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA.
| | - Melanie C Hilman
- Department of Neurosurgery, Center for Brain Injury & Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Feikun Yang
- Department of Medicine, Penn Institute for Regenerative Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Musculoskeletal Program, Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wenli Yang
- Department of Medicine, Penn Institute for Regenerative Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - D Kacy Cullen
- Department of Neurosurgery, Center for Brain Injury & Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA.
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Musculoskeletal Program, Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Essouma M. Autoimmune inflammatory myopathy biomarkers. Clin Chim Acta 2024; 553:117742. [PMID: 38176522 DOI: 10.1016/j.cca.2023.117742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The autoimmune inflammatory myopathy disease spectrum, commonly known as myositis, is a group of systemic diseases that mainly affect the muscles, skin and lungs. Biomarker assessment helps in understanding disease mechanisms, allowing for the implementation of precise strategies in the classification, diagnosis, and management of these diseases. This review examines the pathogenic mechanisms and highlights current data on blood and tissue biomarkers of autoimmune inflammatory myopathies.
Collapse
Affiliation(s)
- Mickael Essouma
- Network of Immunity in Infections, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Cameroon
| |
Collapse
|
12
|
Robaszkiewicz K, Siatkowska M, Wadman RI, Kamsteeg EJ, Chen Z, Merve A, Parton M, Bugiardini E, de Bie C, Moraczewska J. A Novel Variant in TPM3 Causing Muscle Weakness and Concomitant Hypercontractile Phenotype. Int J Mol Sci 2023; 24:16147. [PMID: 38003336 PMCID: PMC10671854 DOI: 10.3390/ijms242216147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
A novel variant of unknown significance c.8A > G (p.Glu3Gly) in TPM3 was detected in two unrelated families. TPM3 encodes the transcript variant Tpm3.12 (NM_152263.4), the tropomyosin isoform specifically expressed in slow skeletal muscle fibers. The patients presented with slowly progressive muscle weakness associated with Achilles tendon contractures of early childhood onset. Histopathology revealed features consistent with a nemaline rod myopathy. Biochemical in vitro assays performed with reconstituted thin filaments revealed defects in the assembly of the thin filament and regulation of actin-myosin interactions. The substitution p.Glu3Gly increased polymerization of Tpm3.12, but did not significantly change its affinity to actin alone. Affinity of Tpm3.12 to actin in the presence of troponin ± Ca2+ was decreased by the mutation, which was due to reduced interactions with troponin. Altered molecular interactions affected Ca2+-dependent regulation of the thin filament interactions with myosin, resulting in increased Ca2+ sensitivity and decreased relaxation of the actin-activated myosin ATPase activity. The hypercontractile molecular phenotype probably explains the distal joint contractions observed in the patients, but additional research is needed to explain the relatively mild severity of the contractures. The slowly progressive muscle weakness is most likely caused by the lack of relaxation and prolonged contractions which cause muscle wasting. This work provides evidence for the pathogenicity of the TPM3 c.8A > G variant, which allows for its classification as (likely) pathogenic.
Collapse
Affiliation(s)
- Katarzyna Robaszkiewicz
- Department of Biochemistry and Cell Biology, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (K.R.); (M.S.)
| | - Małgorzata Siatkowska
- Department of Biochemistry and Cell Biology, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (K.R.); (M.S.)
| | - Renske I. Wadman
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Erik-Jan Kamsteeg
- Department of Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Zhiyong Chen
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, The National Hospital for Neurology, London WC1N 3BG, UK; (Z.C.); (M.P.); (E.B.)
- Department of Neurology, National Neuroscience Institute, Singapore 308433, Singapore
| | - Ashirwad Merve
- Department of Neuropathology, UCL Queen Square Institute of Neurology, The National Hospital for Neurology, London WC1N 3BG, UK;
| | - Matthew Parton
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, The National Hospital for Neurology, London WC1N 3BG, UK; (Z.C.); (M.P.); (E.B.)
| | - Enrico Bugiardini
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, The National Hospital for Neurology, London WC1N 3BG, UK; (Z.C.); (M.P.); (E.B.)
| | - Charlotte de Bie
- Department of Genetics, University Medical Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Joanna Moraczewska
- Department of Biochemistry and Cell Biology, Kazimierz Wielki University, 85-671 Bydgoszcz, Poland; (K.R.); (M.S.)
| |
Collapse
|
13
|
Habibian JS, Bolino M, Qian A, Woolsey R, Quilici D, Petereit J, Ferguson BS. Class I HDAC inhibitors attenuate dexamethasone-induced muscle atrophy via increased protein kinase C (PKC) delta phosphorylation. Cell Signal 2023; 110:110815. [PMID: 37478958 PMCID: PMC10528066 DOI: 10.1016/j.cellsig.2023.110815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Skeletal muscle atrophy is defined by wasting or decrease in muscle mass owing to injury, aging, malnutrition, chronic disuse, or physical consequences of chronic illness. Under normal physiological conditions, a network of signal transduction pathways serves to balance muscle protein synthesis and proteolysis; however, metabolic shifts occur from protein synthesis to protein degradation that leads to a reduction in cross-sectional myofibers and can result in loss of skeletal muscle mass (atrophy) over time. Recent evidence highlights posttranslational modifications (PTMs) such as acetylation and phosphorylation in contractile dysfunction and muscle wasting. Indeed, histone deacetylase (HDAC) inhibitors have been shown to attenuate muscle atrophy and delay muscle damage in response to nutrient deprivation, in models of metabolic dysfunction and genetic models of muscle disease (e.g., muscle dystrophy). Despite our current understanding of lysine acetylation in muscle physiology, a role for HDACs in the regulation of muscle signal transduction remains a 'black box.' Using C2C12 myotubes stimulated with dexamethasone (Dex) as a model of muscle atrophy, we report that protein kinase C delta (PKCδ) phosphorylation decreased at threonine 505 (T505) and serine 643 (S643) in myotubes in response to muscle atrophy; these residues are important for PKCδ activity. Interestingly, PKCδ phosphorylation was restored/increased in myotubes treated with a pan-HDAC inhibitor or a class I selective HDAC inhibitor targeting HDACs1, -2, and - 3 in response to Dex. Moreover, we observed that Dex induced atrophy in skeletal muscle tissue in mice; this reduction in atrophy occurred rapidly, with weight loss noted by day 3 post-Dex and muscle weight loss noted by day 7. Similar to our findings in C2C12 myotubes, Dex attenuated phosphorylation of PKCδ at S643, while HDAC inhibition restored or increased PKCδ phosphorylation at both T505 and S643 in the tibialis anterior. Consistent with this hypothesis, we report that HDAC inhibition could not restore myotube size in response to Dex in the presence of a PKCδ inhibitor or when overexpressing a dominant negative PKCδ. Additionally, the overexpression of a constitutively active PKCδ prevented Dex-induced myotube atrophy. Combined, these data suggest that HDACs regulate muscle physiology via changes in intracellular signaling, namely PKCδ phosphorylation. Whether HDACs regulate PKCδ through canonical (e.g. gene-mediated regulation of phosphatases) or non-canonical (e.g. direct deacetylation of PKCδ to change phosphorylation states) mechanisms remain unclear and future research is needed to clarify this point.
Collapse
Affiliation(s)
- Justine S Habibian
- Department of Nutrition, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Pharmacology and Physiology, The University of Nevada Reno, Reno, NV 89557, USA
| | - Matthew Bolino
- Department of Nutrition, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV 89557, United States of America
| | - Anthony Qian
- Cellular and Molecular Pharmacology and Physiology, The University of Nevada Reno, Reno, NV 89557, USA
| | - Rebekah Woolsey
- Mick Hitchcock, Ph.D. Nevada Proteomics Center (RRID:SCR_017761), The University of Nevada Reno, Reno, NV 89557, USA
| | - David Quilici
- Mick Hitchcock, Ph.D. Nevada Proteomics Center (RRID:SCR_017761), The University of Nevada Reno, Reno, NV 89557, USA
| | - Juli Petereit
- Nevada Bioinformatics Center (RRID:SCR_017802), The University of Nevada Reno, Reno, NV 89557, USA
| | - Bradley S Ferguson
- Department of Nutrition, The University of Nevada Reno, Reno, NV 89557, United States of America; Cellular and Molecular Biology, The University of Nevada Reno, Reno, NV 89557, United States of America; Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, The University of Nevada Reno, Reno, NV 89557, United States of America.
| |
Collapse
|
14
|
Suárez-Calvet X, Fernández-Simón E, Natera D, Jou C, Pinol-Jurado P, Villalobos E, Ortez C, Monceau A, Schiava M, Codina A, Verdu-Díaz J, Clark J, Laidler Z, Mehra P, Gokul-Nath R, Alonso-Perez J, Marini-Bettolo C, Tasca G, Straub V, Guglieri M, Nascimento A, Diaz-Manera J. Decoding the transcriptome of Duchenne muscular dystrophy to the single nuclei level reveals clinical-genetic correlations. Cell Death Dis 2023; 14:596. [PMID: 37673877 PMCID: PMC10482944 DOI: 10.1038/s41419-023-06103-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
Duchenne muscular dystrophy is a genetic disease produced by mutations in the dystrophin gene characterized by early onset muscle weakness leading to severe and irreversible disability. The cellular and molecular consequences of the lack of dystrophin in humans are only partially known, which is crucial for the development of new therapies aiming to slow or stop the progression of the disease. Here we have analyzed quadriceps muscle biopsies of seven DMD patients aged 2 to 4 years old and five age and gender matched controls using single nuclei RNA sequencing (snRNAseq) and correlated the results obtained with clinical data. SnRNAseq identified significant differences in the proportion of cell population present in the muscle samples, including an increase in the number of regenerative fibers, satellite cells, and fibro-adipogenic progenitor cells (FAPs) and a decrease in the number of slow fibers and smooth muscle cells. Muscle samples from the younger patients with stable mild weakness were characterized by an increase in regenerative fibers, while older patients with moderate and progressive weakness were characterized by loss of muscle fibers and an increase in FAPs. An analysis of the gene expression profile in muscle fibers identified a strong regenerative signature in DMD samples characterized by the upregulation of genes involved in myogenesis and muscle hypertrophy. In the case of FAPs, we observed upregulation of genes involved in the extracellular matrix regeneration but also several signaling pathways. Indeed, further analysis of the potential intercellular communication profile showed a dysregulation of the communication profile in DMD samples identifying FAPs as a key regulator of cell signaling in DMD muscle samples. In conclusion, our study has identified significant differences at the cellular and molecular levels in the different cell populations present in skeletal muscle samples of patients with DMD compared to controls.
Collapse
Affiliation(s)
- Xavier Suárez-Calvet
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041, Barcelona, Spain
| | - Esther Fernández-Simón
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Daniel Natera
- Neuromuscular Disorders Unit. Neurology department, Hospital Sant Joan de Deu, Esplugues de Llobregat, Spain
| | - Cristina Jou
- Pathology department, Hospital Sant Joan de Deu, Esplugues de Llobregat, Spain
| | - Patricia Pinol-Jurado
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Elisa Villalobos
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Carlos Ortez
- Neuromuscular Disorders Unit. Neurology department, Hospital Sant Joan de Deu, Esplugues de Llobregat, Spain
| | - Alexandra Monceau
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Marianela Schiava
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Anna Codina
- Pathology department, Hospital Sant Joan de Deu, Esplugues de Llobregat, Spain
| | - José Verdu-Díaz
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - James Clark
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Zoe Laidler
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Priyanka Mehra
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Rasya Gokul-Nath
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Jorge Alonso-Perez
- Neuromuscular Disease Unit. Neurology Department. Hospital Nuestra Señora de Candelaria, Fundación Canaria Instituto de Investigación Sanitaria de Canarias (FIISC), Tenerife, Spain
| | - Chiara Marini-Bettolo
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Giorgio Tasca
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Volker Straub
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK
| | - Andrés Nascimento
- Neuromuscular Disorders Unit. Neurology department, Hospital Sant Joan de Deu, Esplugues de Llobregat, Spain
| | - Jordi Diaz-Manera
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041, Barcelona, Spain.
- John Walton Muscular Dystrophy Research Center, Newcastle University Translational and Clinical Research Institute, Newcastle Upon Tyne, UK.
| |
Collapse
|
15
|
Brockbals L, Garrett-Rickman S, Fu S, Ueland M, McNevin D, Padula MP. Estimating the time of human decomposition based on skeletal muscle biopsy samples utilizing an untargeted LC-MS/MS-based proteomics approach. Anal Bioanal Chem 2023; 415:5487-5498. [PMID: 37423904 PMCID: PMC10444689 DOI: 10.1007/s00216-023-04822-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
Accurate estimation of the postmortem interval (PMI) is crucial in forensic medico-legal investigations to understand case circumstances (e.g. narrowing down list of missing persons or include/exclude suspects). Due to the complex decomposition chemistry, estimation of PMI remains challenging and currently often relies on the subjective visual assessment of gross morphological/taphonomic changes of a body during decomposition or entomological data. The aim of the current study was to investigate the human decomposition process up to 3 months after death and propose novel time-dependent biomarkers (peptide ratios) for the estimation of decomposition time. An untargeted liquid chromatography tandem mass spectrometry-based bottom-up proteomics workflow (ion mobility separated) was utilized to analyse skeletal muscle, collected repeatedly from nine body donors decomposing in an open eucalypt woodland environment in Australia. Additionally, general analytical considerations for large-scale proteomics studies for PMI determination are raised and discussed. Multiple peptide ratios (human origin) were successfully proposed (subgroups < 200 accumulated degree days (ADD), < 655 ADD and < 1535 ADD) as a first step towards generalised, objective biochemical estimation of decomposition time. Furthermore, peptide ratios for donor-specific intrinsic factors (sex and body mass) were found. Search of peptide data against a bacterial database did not yield any results most likely due to the low abundance of bacterial proteins within the collected human biopsy samples. For comprehensive time-dependent modelling, increased donor number would be necessary along with targeted confirmation of proposed peptides. Overall, the presented results provide valuable information that aid in the understanding and estimation of the human decomposition processes.
Collapse
Affiliation(s)
- Lana Brockbals
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia
| | - Samara Garrett-Rickman
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia
| | - Shanlin Fu
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia
| | - Maiken Ueland
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia
| | - Dennis McNevin
- Centre for Forensic Science, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia
| | - Matthew P Padula
- School of Life Sciences, Faculty of Science, University of Technology Sydney, PO Box 123, Broadway, NSW, 2007, Australia.
| |
Collapse
|
16
|
Chan AHP, Jain I, Oropeza BP, Zhou T, Nelsen B, Geisse NA, Huang NF. Combinatorial extracellular matrix cues with mechanical strain induce differential effects on myogenesis in vitro. Biomater Sci 2023; 11:5893-5907. [PMID: 37477446 PMCID: PMC10443049 DOI: 10.1039/d3bm00448a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/12/2023] [Indexed: 07/22/2023]
Abstract
Skeletal muscle regeneration remains a clinical unmet need for volumetric muscle loss and atrophy where muscle function cannot be restored to prior capacity. Current experimental approaches do not account for the complex microenvironmental factors that modulate myogenesis. In this study we developed a biomimetic tissue chip platform to systematically study the combined effects of the extracellular matrix (ECM) microenvironment and mechanical strain on myogenesis of murine myoblasts. Using stretchable tissue chips composed of collagen I (C), fibronectin (F) and laminin (L), as well as their combinations thereof, we tested the addition of mechanical strain regimens on myogenesis at the transcriptomic and translational levels. Our results show that ECMs have a significant effect on myotube formation in C2C12 murine myoblasts. Under static conditions, laminin substrates induced the longest myotubes, whereas fibronectin produced the widest myotubes. Combinatorial ECMs showed non-intuitive effects on myotube formation. Genome-wide analysis revealed the upregulation in actin cytoskeletal related genes that are suggestive of myogenesis. When mechanical strain was introduced to C + F + L combinatorial ECM substrates in the form of constant or intermittent uniaxial strain at low (5%) and high (15%) levels, we observed synergistic enhancements in myotube width, along with transcriptomic upregulation in myosin heavy chain genes. Together, these studies highlight the complex role of microenvironmental factors such as ECM interactions and strain on myotube formation and the underlying signaling pathways.
Collapse
Affiliation(s)
- Alex H P Chan
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ishita Jain
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Beu P Oropeza
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Tony Zhou
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | | | | | - Ngan F Huang
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
17
|
Li S, Lin S, Jiang P, Feng Q, Bao Z, Huang Y, Sun N. Discovery of Potential Protein Markers Associated with Quality Characteristics of Antarctic Krill ( Euphausia superba) Surimi Gel. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37478023 DOI: 10.1021/acs.jafc.3c02860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Antarctic krill are a consumption resource with great exploitation potential. However, the poor gel properties of Antarctic krill meat seriously limit its high-value application. In the present study, the quality characteristics and proteome changes of the κ-/ι-carrageenan-Antarctic krill surimi gel were systematically analyzed and compared. In addition, the transcriptome sequencing of Antarctic krill was carried out, which filled the gap in the Antarctic krill database. Higher molecular forces (disulfide bond and hydrophobic interaction) and the degree of network cross-linking significantly promoted the formation of κ/ι-carrageenan-Antarctic krill surimi compared to that of Antarctic krill surimi. This is the first study to investigate and map potential protein markers for quality characteristics of Antarctic krill surimi based on mass spectrometry-based label-free quantitative proteomics. The results could provide a theoretical reference for the quality control of Antarctic krill during application.
Collapse
Affiliation(s)
- Shuang Li
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Songyi Lin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Pengfei Jiang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Qi Feng
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Zhijie Bao
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Yihan Huang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Na Sun
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
18
|
Yin Y, Zhang Y, Hua Z, Wu A, Pan X, Yang J, Wang X. Muscle transcriptome analysis provides new insights into the growth gap between fast- and slow-growing Sinocyclocheilus grahami. Front Genet 2023; 14:1217952. [PMID: 37538358 PMCID: PMC10394708 DOI: 10.3389/fgene.2023.1217952] [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: 05/06/2023] [Accepted: 07/06/2023] [Indexed: 08/05/2023] Open
Abstract
Sinocyclocheilus grahami is an economically valuable and famous fish in Yunnan Province, China. However, given its slow growth (40 g/2 years) and large growth differences among individuals, its growth performance needs to be improved for sustainable future use, in which molecular breeding technology can play an important role. In the current study, we conducted muscle transcriptomic analysis to investigate the growth gaps among individuals and the mechanism underlying growth within 14 fast- and 14 slow-growth S. grahami. In total, 1,647 differentially expressed genes (DEGs) were obtained, including 947 up-regulated and 700 down-regulated DEGs in fast-growth group. Most DEGs were significantly enriched in ECM-receptor interaction, starch and sucrose metabolism, glycolysis/gluconeogenesis, pyruvate metabolism, amino acids biosynthesis and metabolism, peroxisome, and PPAR signaling pathway. Some genes related to glycogen degradation, glucose transport, and glycolysis (e.g., adipoq, prkag1, slc2a1, agl, pygm, pgm1, pfkm, gapdh, aldoa, pgk1, pgam2, bpgm, and eno3) were up-regulated, while some genes related to fatty acid degradation and transport (e.g., acox1, acaa1, fabp1b.1, slc27a1, and slc27a2) and amino acid metabolism (e.g., agxt, shmt1, glula, and cth) were down-regulated in the fast-growth group. Weighted gene co-expression network analysis identified col1a1, col1a2, col5a1, col6a2, col10a1, col26a1, bglap, and krt15 as crucial genes for S. grahami growth. Several genes related to bone and muscle growth (e.g., bmp2, bmp3, tgfb1, tgfb2, gdf10, and myog) were also up-regulated in the fast-growth group. These results suggest that fast-growth fish may uptake adequate energy (e.g., glucose, fatty acid, and amino acids) from fodder, with excess energy substances used to synthesize collagen to accelerate bone and muscle growth after normal life activities are maintained. Moreover, energy uptake may be the root cause, while collagen synthesis may be the direct reason for the growth gap between fast- and slow-growth fish. Hence, improving food intake and collagen synthesis may be crucial for accelerating S. grahami growth, and further research is required to fully understand and confirm these associations.
Collapse
Affiliation(s)
- Yanhui Yin
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yuanwei Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zexiang Hua
- Fishery Technology Extension Station of Yunnan, Kunming, Yunnan, China
| | - Anli Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaofu Pan
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Junxing Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaoai Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Innovative Academy of Seed Design, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Plateau Fish Breeding, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Engineering Research Center for Plateau-Lake Health and Restoration, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| |
Collapse
|
19
|
Luo X, Zhang H, Cao X, Yang D, Yan Y, Lu J, Wang X, Wang H. Endurance Exercise-Induced Fgf21 Promotes Skeletal Muscle Fiber Conversion through TGF-β1 and p38 MAPK Signaling Pathway. Int J Mol Sci 2023; 24:11401. [PMID: 37511159 PMCID: PMC10379449 DOI: 10.3390/ijms241411401] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Fgf21 has been identified as playing a regulatory role in muscle growth and function. Although the mechanisms through which endurance training regulates skeletal muscle have been widely studied, the contribution of Fgf21 remains poorly understood. Here, muscle size and function were measured, and markers of fiber type were evaluated using immunohistochemistry, immunoblots, or qPCR in endurance-exercise-trained wild-type and Fgf21 KO mice. We also investigated Fgf21-induced fiber conversion in C2C12 cells, which were incubated with lentivirus and/or pathway inhibitors. We found that endurance exercise training enhanced the Fgf21 levels of liver and GAS muscle and exercise capacity and decreased the distribution of skeletal muscle fiber size, and fast-twitch fibers were observed converting to slow-twitch fibers in the GAS muscle of mice. Fgf21 promoted the markers of fiber-type transition and eMyHC-positive myotubes by inhibiting the TGF-β1 signaling axis and activating the p38 MAPK signaling pathway without apparent crosstalk. Our findings suggest that the transformation and function of skeletal muscle fiber types in response to endurance training could be mediated by Fgf21 and its downstream signaling pathways. Our results illuminate the mechanisms of Fgf21 in endurance-exercise-induced fiber-type conversion and suggest a potential use of Fgf21 in improving muscle health and combating fatigue.
Collapse
Affiliation(s)
- Xiaomao Luo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Huiling Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xiaorui Cao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Ding Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Jiayin Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Xiaonan Wang
- Renal Division, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| |
Collapse
|
20
|
Saad MK, Yuen JSK, Joyce CM, Li X, Lim T, Wolfson TL, Wu J, Laird J, Vissapragada S, Calkins OP, Ali A, Kaplan DL. Continuous fish muscle cell line with capacity for myogenic and adipogenic-like phenotypes. Sci Rep 2023; 13:5098. [PMID: 36991012 PMCID: PMC10060565 DOI: 10.1038/s41598-023-31822-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Cell-cultivated fish offers the potential for a more ethical, sustainable, and safe seafood system. However, fish cell culture is relatively understudied in comparison to mammalian cells. Here, we established and characterized a continuous Atlantic mackerel (Scomber scombrus) skeletal muscle cell line ("Mack" cells). The cells were isolated from muscle biopsies of fresh-caught fish, with separate isolations performed from two distinct fish. Mack1 cells (cells from the first isolation) were cultured for over a year and subcultured over 130 times. The cells proliferated at initial doubling times of 63.9 h (± 19.1 SD). After a spontaneous immortalization crisis from passages 37-43, the cells proliferated at doubling times of 24.3 h (± 4.91 SD). A muscle phenotype was confirmed through characterization of muscle stemness and differentiation via paired-box protein 7 and myosin heavy chain immunostaining, respectively. An adipocyte-like phenotype was also demonstrated for the cells through lipid accumulation, confirmed via Oil Red O staining and quantification of neutral lipids. New qPCR primers (HPRT, PAX3B, MYOD1, MYOG, TNNT3A, and PPARG) were tailored to the mackerel genome and used to characterize mackerel cell genotypes. This work provides the first spontaneously immortalized fish muscle cell line for research, ideally serving as a reference for subsequent investigation.
Collapse
Affiliation(s)
- Michael K Saad
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - John S K Yuen
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Connor M Joyce
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Xinxin Li
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Taehwan Lim
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Talia L Wolfson
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Justin Wu
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Jason Laird
- Research Technology, Tufts University, 16 Dearborn Rd, Somerville, MA, 02144, USA
| | - Sanjana Vissapragada
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Olivia P Calkins
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - Adham Ali
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA
| | - David L Kaplan
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, 4 Colby St, Medford, MA, 02155, USA.
| |
Collapse
|
21
|
Kuntawala DH, Martins F, Vitorino R, Rebelo S. Automatic Text-Mining Approach to Identify Molecular Target Candidates Associated with Metabolic Processes for Myotonic Dystrophy Type 1. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2283. [PMID: 36767649 PMCID: PMC9915907 DOI: 10.3390/ijerph20032283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant hereditary disease caused by abnormal expansion of unstable CTG repeats in the 3' untranslated region of the myotonic dystrophy protein kinase (DMPK) gene. This disease mainly affects skeletal muscle, resulting in myotonia, progressive distal muscle weakness, and atrophy, but also affects other tissues and systems, such as the heart and central nervous system. Despite some studies reporting therapeutic strategies for DM1, many issues remain unsolved, such as the contribution of metabolic and mitochondrial dysfunctions to DM1 pathogenesis. Therefore, it is crucial to identify molecular target candidates associated with metabolic processes for DM1. In this study, resorting to a bibliometric analysis, articles combining DM1, and metabolic/metabolism terms were identified and further analyzed using an unbiased strategy of automatic text mining with VOSviewer software. A list of candidate molecular targets for DM1 associated with metabolic/metabolism was generated and compared with genes previously associated with DM1 in the DisGeNET database. Furthermore, g:Profiler was used to perform a functional enrichment analysis using the Gene Ontology (GO) and REAC databases. Enriched signaling pathways were identified using integrated bioinformatics enrichment analyses. The results revealed that only 15 of the genes identified in the bibliometric analysis were previously associated with DM1 in the DisGeNET database. Of note, we identified 71 genes not previously associated with DM1, which are of particular interest and should be further explored. The functional enrichment analysis of these genes revealed that regulation of cellular metabolic and metabolic processes were the most associated biological processes. Additionally, a number of signaling pathways were found to be enriched, e.g., signaling by receptor tyrosine kinases, signaling by NRTK1 (TRKA), TRKA activation by NGF, PI3K-AKT activation, prolonged ERK activation events, and axon guidance. Overall, several valuable target candidates related to metabolic processes for DM1 were identified, such as NGF, NTRK1, RhoA, ROCK1, ROCK2, DAG, ACTA, ID1, ID2 MYOD, and MYOG. Therefore, our study strengthens the hypothesis that metabolic dysfunctions contribute to DM1 pathogenesis, and the exploitation of metabolic dysfunction targets is crucial for the development of future therapeutic interventions for DM1.
Collapse
|
22
|
Dowling P, Gargan S, Swandulla D, Ohlendieck K. Fiber-Type Shifting in Sarcopenia of Old Age: Proteomic Profiling of the Contractile Apparatus of Skeletal Muscles. Int J Mol Sci 2023; 24:2415. [PMID: 36768735 PMCID: PMC9916839 DOI: 10.3390/ijms24032415] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The progressive loss of skeletal muscle mass and concomitant reduction in contractile strength plays a central role in frailty syndrome. Age-related neuronal impairments are closely associated with sarcopenia in the elderly, which is characterized by severe muscular atrophy that can considerably lessen the overall quality of life at old age. Mass-spectrometry-based proteomic surveys of senescent human skeletal muscles, as well as animal models of sarcopenia, have decisively improved our understanding of the molecular and cellular consequences of muscular atrophy and associated fiber-type shifting during aging. This review outlines the mass spectrometric identification of proteome-wide changes in atrophying skeletal muscles, with a focus on contractile proteins as potential markers of changes in fiber-type distribution patterns. The observed trend of fast-to-slow transitions in individual human skeletal muscles during the aging process is most likely linked to a preferential susceptibility of fast-twitching muscle fibers to muscular atrophy. Studies with senescent animal models, including mostly aged rodent skeletal muscles, have confirmed fiber-type shifting. The proteomic analysis of fast versus slow isoforms of key contractile proteins, such as myosin heavy chains, myosin light chains, actins, troponins and tropomyosins, suggests them as suitable bioanalytical tools of fiber-type transitions during aging.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, D53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
23
|
Joyce W, Ripley DM, Gillis T, Black AC, Shiels HA, Hoffmann FG. A Revised Perspective on the Evolution of Troponin I and Troponin T Gene Families in Vertebrates. Genome Biol Evol 2022; 15:6904147. [PMID: 36518048 PMCID: PMC9825255 DOI: 10.1093/gbe/evac173] [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: 05/05/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The troponin (Tn) complex, responsible for the Ca2+ activation of striated muscle, is composed of three interacting protein subunits: TnC, TnI, and TnT, encoded by TNNC, TNNI, and TNNT genes. TNNI and TNNT are sister gene families, and in mammals the three TNNI paralogs (TNNI1, TNNI2, TNNI3), which encode proteins with tissue-specific expression, are each in close genomic proximity with one of the three TNNT paralogs (TNNT2, TNNT3, TNNT1, respectively). It has been widely presumed that all vertebrates broadly possess genes of these same three classes, although earlier work has overlooked jawless fishes (cyclostomes) and cartilaginous fishes (chimeras, rays, and sharks), which are distantly related to other jawed vertebrates. With a new phylogenetic and synteny analysis of a diverse array of vertebrates including these taxonomic groups, we define five distinct TNNI classes (TNNI1-5), with TNNI4 and TNNI5 being only present in non-amniote vertebrates and typically found in tandem, and four classes of TNNT (TNNT1-4). These genes are located in four genomic loci that were generated by the 2R whole-genome duplications. TNNI3, encoding "cardiac TnI" in tetrapods, was independently lost in cartilaginous and ray-finned fishes. Instead, ray-finned fishes predominantly express TNNI1 in the heart. TNNI5 is highly expressed in shark hearts and contains a N-terminal extension similar to that of TNNI3 found in tetrapod hearts. Given that TNNI3 and TNNI5 are distantly related, this supports the hypothesis that the N-terminal extension may be an ancestral feature of vertebrate TNNI and not an innovation unique to TNNI3, as has been commonly believed.
Collapse
Affiliation(s)
| | - Daniel M Ripley
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Todd Gillis
- Department of Integrative Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Amanda Coward Black
- Department of Biochemistry, Molecular Biology, Entomology, and Plant Pathology, Mississippi State University, Starkville, Mississippi 39762, USA
| | - Holly A Shiels
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | | |
Collapse
|
24
|
Hendin N, Gordon T, Shenkar N, Wurtzel O. Molecular characterization of the immediate wound response of the solitary ascidian Polycarpa mytiligera. Dev Dyn 2022; 251:1968-1981. [PMID: 36001356 PMCID: PMC10087333 DOI: 10.1002/dvdy.526] [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: 03/13/2022] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Injury response is key to successful regeneration. Yet, transcriptome analyses of injury response were performed only on a handful of regenerative organisms. Here, we studied the injury response of the solitary ascidian Polycarpa mytiligera, an emerging model system, capable of regenerating any body part. We used the siphon as a model for studying transcriptional changes following injury, and identified genes that were activated in the initial 24 hours post amputation (hpa). RESULTS Highly conserved genes, such as bone morphogenetic protein-1 (BMP1), growth hormone secretagogue receptor (GHSR) and IL-17, were upregulated by 12 hpa, yet their expression was sustained only in non-regenerating tissue fragments. We optimized fluorescent in situ hybridization, and found that the majority of BMP1+ cells were localized to the rigid tunic that covers the animal. This highlights the importance of this tissue, particularly during injury response. BMP1 was overexpressed following injuries to other body regions, suggesting that it was a part of a common injury-induced program. CONCLUSION Our study suggests that, initially, specific injury-induced genes were upregulated in P. mytiligera organs, yet, later, a unique transcriptional profile was observed only in regenerating tissues. These findings highlight the importance of studying diverse regenerating and non-regenerating organisms for complete understanding of regeneration.
Collapse
Affiliation(s)
- Noam Hendin
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Tal Gordon
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
| | - Noa Shenkar
- School of Zoology, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- The Steinhardt Museum of Natural History, Israel National Center for Biodiversity StudiesTel‐Aviv UniversityTel‐AvivIsrael
| | - Omri Wurtzel
- The School of Neurobiology, Biochemistry & Biophysics, George S. Wise Faculty of Life SciencesTel Aviv UniversityTel AvivIsrael
- Sagol School of NeuroscienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
25
|
Leite L, Matos P, Leon-Justel A, Espírito-Santo C, Rodríguez-Padial L, Rodrigues F, Orozco D, Redon J. High sensitivity troponins: A potential biomarkers of cardiovascular risk for primary prevention. Front Cardiovasc Med 2022; 9:1054959. [PMID: 36531726 PMCID: PMC9748104 DOI: 10.3389/fcvm.2022.1054959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/14/2022] [Indexed: 03/07/2024] Open
Abstract
There have been several approaches to building charts for CV risk, all of which have both strengths and limitations. Identifying early organ damage provides relevant information and should be included in risk charts, although the direct relationship with risk is imprecise, variability between operators at the time to assess, and low availability in some healthcare systems, limits its use. Biomarkers, like troponin (cTns) isoforms cTnI and cTnT, a cardiac specific myocyte injury marker, have the great advantage of being relatively reproducible, more readily accessible, and applicable to different populations. New and improved troponin assays have good analytical performance, can measure very low levels of circulating troponin, and have low intra individual variation, below 10 %. Several studies have analyzed the blood levels in healthy subjects and their predictive value for cardiovascular events in observational, prospective and post-hoc studies. All of them offered relevant information and shown that high sensitivity hs-cTnI has a place as an additional clinical marker to add to current charts, and it also reflects sex- and age-dependent differences. Although few more questions need to be answered before recommend cTnI for assessing CV risk in primary prevention, seems to be a potential strong marker to complement CV risk charts.
Collapse
Affiliation(s)
- Luis Leite
- Cardiology Department, Coimbra University Hospital, University of Coimbra, Coimbra, Portugal
| | - Pedro Matos
- APDP e Hospital CUF Infante Santo, Lisbon, Portugal
| | - Antonio Leon-Justel
- Department of Laboratory Medicine, Virgen Macarena University Hospital, Seville, Spain
| | | | | | | | - Domingo Orozco
- Department of Clinical Medicine, Miguel Hernández University, Elche, Spain
| | - Josep Redon
- INCLIVA Research Institute, University of Valencia, Valencia, Spain
- CIBERObn Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
26
|
Kłaczewska J, Tomasiuk R. Cardiac Troponin T in Neonates from Normal and Gestational Diabetes Mellitus Pregnancy. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6171687. [PMID: 36303588 PMCID: PMC9596251 DOI: 10.1155/2022/6171687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022]
Abstract
This study is aimed at testing the hypothesis that serum analysis of high-sensitivity troponin T in neonates may serve as a diagnostic tool to predict the risk of gestational diabetes mellitus (GDM). 86 newborns were studied and stratified into two groups: healthy group; newborns with body weight ≥ 10th percentile, born in good condition (APG 8-10pts) of pregnancy not complicated by diabetes, and the GDM group; neonates born to mothers with type 1 or type 2 diabetes. Results. The study revealed minimal troponin levels in GDM, and healthy groups equal to 0.02 ng/mL and 0.028 ng/mL, respectively. The GDM group is defined by an interquartile range of hs-TnT higher than the healthy group. This study confirms previously reported upper levels of troponin in healthy children. There are possible health problems that can appear during infancy and influence the further development of a child affected by GDM.
Collapse
Affiliation(s)
- Joanna Kłaczewska
- Józef Polikarp Brudzinski Children's Clinical Hospital in Warsaw, 63A Żwirki i Wigury St., 02-91 Warsaw, Poland
| | - Ryszard Tomasiuk
- Kazimierz Pulaski University of Technology and Humanities Radom, Faculty of Medical Sciences and Health Sciences, Radom, Poland
| |
Collapse
|
27
|
Crum RJ, Johnson SA, Jiang P, Jui JH, Zamora R, Cortes D, Kulkarni M, Prabahar A, Bolin J, Gann E, Elster E, Schobel SA, Larie D, Cockrell C, An G, Brown B, Hauskrecht M, Vodovotz Y, Badylak SF. Transcriptomic, Proteomic, and Morphologic Characterization of Healing in Volumetric Muscle Loss. Tissue Eng Part A 2022; 28:941-957. [PMID: 36039923 DOI: 10.1089/ten.tea.2022.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Skeletal muscle has a robust, inherent ability to regenerate in response to injury from acute to chronic. In severe trauma, however, complete regeneration is not possible, resulting in a permanent loss of skeletal muscle tissue referred to as volumetric muscle loss (VML). There are few consistently reliable therapeutic or surgical options to address VML. A major limitation in investigation of possible therapies is the absence of a well-characterized large animal model. Here, we present results of a comprehensive transcriptomic, proteomic, and morphologic characterization of wound healing following volumetric muscle loss in a novel canine model of VML which we compare to a nine-patient cohort of combat-associated VML. The canine model is translationally relevant as it provides both a regional (spatial) and temporal map of the wound healing processes that occur in human VML. Collectively, these data show the spatiotemporal transcriptomic, proteomic, and morphologic properties of canine VML healing as a framework and model system applicable to future studies investigating novel therapies for human VML.
Collapse
Affiliation(s)
- Raphael John Crum
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 450 Technology Dr., Suite 300, Pittsburgh, Pennsylvania, United States, 15219;
| | - Scott A Johnson
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, 450 Technology Dr, Suite 300, Pittsburgh, Pennsylvania, United States, 15219;
| | - Peng Jiang
- Cleveland State University, Center for Gene Regulation in Health and Disease, Cleveland, Ohio, United States.,Cleveland State University, Center for Applied Data Analysis and Modeling (ADAM), Cleveland, Ohio, United States.,Cleveland State University, Department of Biological, Geological, and Environmental Sciences (BGES), Cleveland, Ohio, United States;
| | - Jayati H Jui
- University of Pittsburgh, Department of Computer Science, Pittsburgh, Pennsylvania, United States;
| | - Ruben Zamora
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Surgery, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Center for Inflammation and Regeneration Modeling, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Center for Systems Immunology, Pittsburgh, Pennsylvania, United States;
| | - Devin Cortes
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Bioengineering, Pittsburgh, Pennsylvania, United States;
| | - Mangesh Kulkarni
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Bioengineering, Pittsburgh, Pennsylvania, United States;
| | - Archana Prabahar
- Cleveland State University, Center for Gene Regulation in Health and Disease, Cleveland, Ohio, United States;
| | - Jennifer Bolin
- Morgridge Institute for Research, Madison, Wisconsin, United States;
| | - Eric Gann
- Uniformed Services University of the Health Sciences, Surgery, Bethesda, Maryland, United States.,Uniformed Services University of the Health Sciences, Surgical Critical Care Initiative, Department of Surgery, Bethesda, Maryland, United States.,Henry M Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, Maryland, United States;
| | - Eric Elster
- Uniformed Services University of the Health Sciences, Surgery, Bethesda, Maryland, United States.,Henry M Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, Maryland, United States.,Uniformed Services University of the Health Sciences, Surgical Critical Care Initiative, Department of Surgery, Bethesda, Maryland, United States.,Walter Reed Army Medical Center, Bethesda, Maryland, United States;
| | - Seth A Schobel
- Uniformed Services University of the Health Sciences, Surgery, Bethesda, Maryland, United States.,Henry M Jackson Foundation for the Advancement of Military Medicine Inc, Bethesda, Maryland, United States.,Uniformed Services University of the Health Sciences, Surgical Critical Care Initiative, Department of Surgery, Bethesda, Maryland, United States;
| | - Dale Larie
- University of Vermont, Department of Surgery, Burlington, Vermont, United States;
| | - Chase Cockrell
- University of Vermont, Department of Surgery, Burlington, Vermont, United States;
| | - Gary An
- University of Vermont, Department of Surgery, Burlington, Vermont, United States;
| | - Bryan Brown
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Bioengineering, Pittsburgh, Pennsylvania, United States;
| | - Milos Hauskrecht
- University of Pittsburgh, Department of Computer Science, Pittsburgh, Pennsylvania, United States;
| | - Yoram Vodovotz
- University of Pittsburgh, Surgery, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Surgery, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Center for Inflammation and Regeneration Modeling, Pittsburgh, Pennsylvania, United States.,University of Pittsburgh, Center for Systems Immunology, Pittsburgh, Pennsylvania, United States;
| | - Stephen F Badylak
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, United States;
| |
Collapse
|
28
|
Yu J, Loh K, Yang HQ, Du MR, Wu YX, Liao ZY, Guo A, Yang YF, Chen B, Zhao YX, Chen JL, Zhou J, Sun Y, Xiao Q. The Whole-transcriptome Landscape of Diabetes-related Sarcopenia Reveals the Specific Function of Novel lncRNA Gm20743. Commun Biol 2022; 5:774. [PMID: 35915136 PMCID: PMC9343400 DOI: 10.1038/s42003-022-03728-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/15/2022] [Indexed: 11/11/2022] Open
Abstract
While the exact mechanism remains unclear, type 2 diabetes mellitus increases the risk of sarcopenia which is characterized by decreased muscle mass, strength, and function. Whole-transcriptome RNA sequencing and informatics were performed on the diabetes-induced sarcopenia model of db/db mice. To determine the specific function of lncRNA Gm20743, the detection of Mito-Sox, reactive oxygen species, Ethynyl-2′-deoxyuridine, and myosin heavy chain was performed in overexpressed and knockdown-Gm20743 C2C12 cells. RNA-seq data and informatics revealed the key lncRNA-mRNA interactions and indicated a potential regulatory role of lncRNAs. We characterized three core candidate lncRNAs Gm20743, Gm35438, 1700047G03Rik, and their potential function. Furthermore, the results suggested lncRNA Gm20743 may be involved in regulating mitochondrial function, oxidative stress, cell proliferation, and myotube differentiation in skeletal muscle cells. These findings significantly improve our understanding of lncRNAs that may mediate muscle mass, strength, and function in diabetes and represent potential therapeutic targets for diabetes-induced sarcopenia. The role of lncRNA Gm20743 in the development of diabetic sarcopenia is explored using a mouse model.
Collapse
Affiliation(s)
- Jing Yu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kim Loh
- Diabetes & Metabolic Disease Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, VIC, Australia
| | - He-Qin Yang
- Health Outcome Research and Policy, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Meng-Ran Du
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong-Xin Wu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Yin Liao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ai Guo
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-Fei Yang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Chen
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yu-Xing Zhao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin-Liang Chen
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Zhou
- Department of Clinical Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yue Sun
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Xiao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
29
|
Mair J, Giannitsis E, Mills NL, Mueller C. How to deal with unexpected cardiac troponin results. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:e1-e3. [PMID: 35218650 DOI: 10.1093/ehjacc/zuac023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Johannes Mair
- Department of Internal Medicine III-Cardiology and Angiology, Medical University Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | | | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
30
|
Li WY, Cai ZL, Zhang BP, Chen JJ, Ji K. Identification of an immunodominant IgE epitope of Der p 39, a novel allergen of Dermatophagoides pteronyssinus. World Allergy Organ J 2022; 15:100651. [PMID: 35600837 PMCID: PMC9096144 DOI: 10.1016/j.waojou.2022.100651] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/11/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
|
31
|
Zhang X, Chen M, Yan E, Wang Y, Ma C, Zhang P, Yin J. Dietary Malic Acid Supplementation Induces Skeletal Muscle Fiber-Type Transition of Weaned Piglets and Further Improves Meat Quality of Finishing Pigs. Front Nutr 2022; 8:825495. [PMID: 35145985 PMCID: PMC8821922 DOI: 10.3389/fnut.2021.825495] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/27/2021] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to investigate effects of dietary malic acid supplementation on skeletal muscle fiber-type transition during nursery period and the subsequent meat quality of finishing pigs. Results showed that malic acid supplementation for 28 days increased oxidative fiber percentage of weaned piglets, accompanied by the increased aerobic oxidation in serum and longissimus thoracis (LT) muscle. Additionally, activities of total antioxidant capacity and glutathione peroxidase in serum were increased. Moreover, dietary malic acid supplementation during nursery period tended to increase pH24h and significantly decreased drip loss in LT muscle of finishing pigs. The content of total saturated fatty acid (SFA) and total monounsaturated fatty acid in LT muscle was significantly decreased, whereas the ratio of polyunsaturated fatty acid to SFA tended to increase. Together, dietary malic acid supplementation during nursery period can effectively increase antioxidant capacity and oxidative fibers percentage of weaned piglets, and further improve water holding capacity and nutritional values of pork in finishing pigs.
Collapse
Affiliation(s)
- Xin Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Enfa Yan
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yubo Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chenghong Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Pengguang Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Jingdong Yin
| |
Collapse
|