1
|
Huettemeister J, Bögner M, Eggert-Doktor D, Heil E, Primessnig U, Reimers SC, Kirk M, Ramesh G, Nazari-Shafti TZ, Grubitzsch H, Sündermann S, Knosalla C, Zhang K, Falk V, Heinzel FR, Hohendanner F. Structural and functional adaptations of human cardiomyocytes in metabolic disease and heart failure. Am J Physiol Heart Circ Physiol 2025; 328:H1193-H1203. [PMID: 40266293 DOI: 10.1152/ajpheart.00903.2024] [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: 01/06/2025] [Revised: 01/27/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Heart failure (HF), obesity, and diabetes are associated with structural and functional changes that affect the heart at both the organ and cellular levels. Studying isolated adult single cardiomyocytes provides valuable mechanistic insights. However, isolating single cardiomyocytes from human tissue is particularly challenging. This study presents an optimized multiple-step digestion protocol to isolate viable cardiomyocytes from atrial and ventricular human tissue obtained perioperatively or through myocardial biopsies. Using this method and resource, we analyzed calcium-signaling during excitation-contraction coupling and structural features such as t-tubules and mitochondria using confocal microscopy in patients with or without HF, obesity, or diabetes. In a subset of patients undergoing open heart surgery, tissue samples and serum from the great cardiac vein were obtained either under control conditions or upon cardiac volume challenge (VC). We isolated viable cells and observed distinct structural differences between atrial and ventricular cardiomyocytes, including variations in t-tubular and cell size. In atrial cardiomyocytes, when comparing control with patients with HF, the t-tubular networks were unchanged. However, patients with obesity exhibited significantly more t-tubules associated with larger cell sizes. Furthermore, mitochondrial density appeared higher in patients with overweight and diabetes, suggesting that the metabolic status influences cardiomyocyte structure. Finally, when exposing isolated cardiomyocytes with VC serum from the respective patients, excitation-contraction coupling was markedly enhanced, indicating a distention-related alteration of the cardiac secretome with immediate effects on cardiomyocytes. In summary, an optimized protocol for isolating human cardiomyocytes confirmed structural features, identified disease-related changes, and allowed studying the dynamic impact of cardiac distention on secretome-related cardiomyocyte function.NEW & NOTEWORTHY This study presents a novel protocol for isolating human cardiomyocytes, uncovering atrial-ventricular structural differences, obesity-related increases in t-tubules and mitochondria, and metabolic influences on cell architecture. It highlights the dynamic effects of cardiac volume challenge on excitation-contraction coupling through secretome alterations. These advancements provide insights into how conditions like obesity and diabetes reshape cardiomyocyte structure and function, advancing our understanding of heart disease mechanisms.
Collapse
Affiliation(s)
- Judith Huettemeister
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Markus Bögner
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Dirk Eggert-Doktor
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuel Heil
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Primessnig
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie C Reimers
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marzena Kirk
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Girish Ramesh
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Timo Zadegh Nazari-Shafti
- Department of Cardiothoracic and Vascular Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Herko Grubitzsch
- Department of Cardiothoracic and Vascular Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Sündermann
- Department of Cardiothoracic and Vascular Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Knosalla
- Department of Cardiothoracic and Vascular Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kun Zhang
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
| | - Volkmar Falk
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Health Sciences and Technology, Translational Cardiovascular Technologies, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Frank R Heinzel
- Medizinische Klinik, Städtisches Klinikum Dresden, Dresden, Germany
| | - Felix Hohendanner
- Department of Cardiology, Angiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK) Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Zhang C, Tian Y, Liu X, Yang X, Jiang S, Zhang G, Yang C, Liu W, Guo W, Zhao W, Yin D. MiR-495 reverses in the mechanical unloading, random rotating and aging induced muscle atrophy via targeting MyoD and inactivating the Myostatin/TGF-β/Smad3 axis. Arch Biochem Biophys 2025; 764:110273. [PMID: 39701202 DOI: 10.1016/j.abb.2024.110273] [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: 07/31/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
Mechanical unloading can lead to homeostasis imbalance and severe muscle disease, in which muscle atrophy was one of the disused diseases. However, there were limited therapeutic targets for such diseases. In this study, miR-495 was found dramatically reduced in atrophic skeletal muscle induced by mechanical unloading models both in vitro and in vivo, including the random positioning model (RPM), tail-suspension (TS) model, and aged mice model. Enforced miR-495 expression by its mimic could enormously facilitate the differentiation and regeneration of both mouse myoblast C2C12 cells and muscle satellite cells. Furthermore, MyoD was proved as the directly interacted gene of miR-495, and their interaction was crucial for myotube formation. Enforced miR-495 expression could intensively strengthen the muscle mass, in situ muscular electrophysiological indexes, including peak tetanic tension (Po) and peak twitch tension (Pt), and the cross-sectional areas (CSA) of muscle fibers via targeting MyoD and inactivating the Myostatin/TGF-β/Smad3 signaling pathway, indicating that miR-495 can be proposed as an effective target for muscle atrophy treatment induced by in the mechanical unloading, random rotating and aging.
Collapse
Affiliation(s)
- Chenyan Zhang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063, China; Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China.
| | - Yile Tian
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063, China; Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Xinli Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Xuezhou Yang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Shanfeng Jiang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Ge Zhang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063, China; Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Changqing Yang
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Wenjing Liu
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Weihong Guo
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518063, China; Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Wenzhe Zhao
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Dachuan Yin
- Institute for Special Environmental Biophysics, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China.
| |
Collapse
|
3
|
Yin A, Fu Y, Wang T, Li H, Wang X, Ye X, Dong P, Yao W. Fu-Zheng-Li-Fei Recipe (FZLFR) in the treatment of cancer cachexia: Exploration of the efficacy and molecular mechanism based on chemical characterization, experimental research and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118929. [PMID: 39395766 DOI: 10.1016/j.jep.2024.118929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/24/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE FZLFR was derived from a classic traditional Chinese medicine recipe, the Shiquan-Dabu decoction. FZLFR is commonly used in clinical practice to address muscle loss and associated cancer cachexia. However, the mechanism of by which FZLFR acts in cancer cachexia remains unclear. AIM This study aimed to assess the effects and explore the potential mechanism of action of FZLFR in treating cancer cachexia. METHODS Cancer cachexia was induced by inoculating Lewis lung carcinoma cells into the right flank of male C57BL/6 mice. The efficacy of FZLFR was evaluated by comparing changes in body weight, tumor mass, food intake, survival time, weight, and cross-sectional area of the gastrocnemius and anterior tibial muscles. Moreover, inflammatory cytokines, such as TNF-α and IL-6, were detected by ELISA. The chemical components of FZLFR were analyzed using ultra-performance liquid chromatography-coupled with time-of-flight mass spectrometry. Network pharmacology analysis was performed to screen the core targets and potential pathways involved in FZLFR treatment of cancer cachexia. Molecular docking was used to analyze the binding ability of the core targets and key compounds. The expression levels of core targets and targets correlated with skeletal muscle atrophy were also assessed using western blotting. RESULTS FZLFR enhanced the food intake and survival rate of mice with cancer cachexia. It also alleviated tumor-induced body weight loss, tumor growth, and muscle fiber atrophy in these mice. Additionally, it improved the weight and cross-sectional area of the gastrocnemius and anterior tibial muscles. FZLFR down-regulated the serum levels of TNF-α and IL-6. UPLC-ESI-Q-TOF-MS analysis identified 184 compounds in FZLFR. Network pharmacology analysis predicted that TNF signaling pathway, ErbB signaling pathway and VEGF signaling pathway might be essential in FZLFR action. Molecular docking showed that kaempferol, upafolin, apigenin, and luteolin might play key roles in FZLFR treatment. Moreover, FZLFR decreased MAFBx1, MURF1, NF-κB, TWEAK, MAPK8, and EGFR expression levels. FZLFR enhanced the expression of VEGFA and ESR1, as demonstrated by western blotting. CONCLUSIONS FZLFR increased food intake and alleviated muscle atrophy in mice with cancer cachexia. The potential pharmacological mechanisms underlying its anticachexia effects include reducing inflammation, enhancing muscle vascular growth, inhibiting tumor angiogenesis, and modulating estrogen receptors.
Collapse
Affiliation(s)
- Aining Yin
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China; Zhongshan College of Dalian Medical University, Dalian, 116085, China
| | - Yu Fu
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China; Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Tingxin Wang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Honglin Li
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xiufang Wang
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China; Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xueke Ye
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China; Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Peipei Dong
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| | - Wei Yao
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China; Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
4
|
Boeno FP, Roesch LFW, Efron PA, Laitano O. Proteomic Profiling of Hindlimb Skeletal Muscle Disuse in a Murine Model of Sepsis. Crit Care Explor 2024; 6:e1144. [PMID: 39162648 PMCID: PMC11338252 DOI: 10.1097/cce.0000000000001144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024] Open
Abstract
CONTEXT Sepsis leads to multiple organ dysfunction and negatively impacts patient outcomes. Skeletal muscle disuse is a significant comorbidity in septic patients during their ICU stay due to prolonged immobilization. HYPOTHESIS Combination of sepsis and muscle disuse will promote a unique proteomic signature in skeletal muscle in comparison to disuse and sepsis separately. METHODS AND MODELS Following cecal ligation and puncture (CLP) or Sham surgeries, mice were subjected to hindlimb suspension (HLS) or maintained normal ambulation (NA). Tibialis anterior muscles from 24 C57BL6/J male mice were harvested for proteomic analysis. Proteomic profiles were assessed using nano-liquid chromatography with tandem mass spectrometry, followed by data analysis including Partial Least Squares Discriminant Analysis (PLS-DA), to compare the differential protein expression across groups. RESULTS A total of 2876 differentially expressed proteins were identified, with marked differences between groups. In mice subjected to CLP and HLS combined, there was a distinctive proteomic signature characterized by a significant decrease in the expression of proteins involved in mitochondrial function and muscle metabolism, alongside a marked increase in proteins related to muscle degradation pathways. The PLS-DA demonstrated a clear separation among experimental groups, highlighting the unique profile of the CLP/HLS group. This suggests an important interaction between sepsis-induced inflammation and disuse atrophy mechanisms in sepsis-induced myopathy. INTERPRETATIONS AND CONCLUSIONS Our findings reveal a complex proteomic landscape in skeletal muscle exposed to sepsis and disuse, consistent with an exacerbation of muscle protein degradation under these combined stressors. The identified proteins and their roles in cellular stress responses and muscle pathology provide potential targets for intervention to mitigate muscle dysfunction in septic conditions, highlighting the importance of addressing both sepsis and disuse concurrently in clinical and experimental settings.
Collapse
Affiliation(s)
- Franccesco P. Boeno
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL
| | - Luiz Fernando W. Roesch
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL
| | - Philip A. Efron
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, Gainesville, FL
| |
Collapse
|
5
|
Dong X, Su S, Sun Q, Wang P, Hu Q, Wei Q. Aligned Nanofibers Promote Myoblast Polarization and Myogenesis through Activating Rac-Related Signaling Pathways. ACS Biomater Sci Eng 2024; 10:1712-1721. [PMID: 38422457 DOI: 10.1021/acsbiomaterials.4c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The extracellular matrix (ECM) plays a crucial role in regulating cellular behaviors and functions. However, the impact of ECM topography on muscle cell adhesion and differentiation remains poorly understood from a mechanosensing perspective. In this study, we fabricated aligned and random electrospun polycaprolactone (PCL) nanofibers to mimic the structural characteristics of ECM. Mechanism investigations revealed that the orientation of nanofibers promoted C2C12 polarization and myogenesis through Rac-related signaling pathways. Conversely, cells cultured on random fibers exhibited spreading behavior mediated by RhoA/ROCK pathways, resulting in enhanced stress fiber formation but reduced capacity for myogenic differentiation. Our findings highlight the critical role of an ECM structure in muscle regeneration and damage repair, providing novel insights into mechanosensing mechanisms underlying muscle injury diseases.
Collapse
Affiliation(s)
- Xiangyu Dong
- Department of Gynecology and Obstetrics, Frontiers Science Center for Disease-related Molecular Network, West China Second University Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Nephrology, Kidney Research Institute, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Shan Su
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Qian Sun
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Peng Wang
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Qian Hu
- Department of Gynecology and Obstetrics, Frontiers Science Center for Disease-related Molecular Network, West China Second University Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
6
|
Aldahhan RA, Motawei KH, Al-Hariri MT. Lipotoxicity-related sarcopenia: a review. J Med Life 2022; 15:1334-1339. [PMID: 36567835 PMCID: PMC9762358 DOI: 10.25122/jml-2022-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/27/2022] [Indexed: 12/27/2022] Open
Abstract
A body of literature supports the postulation that a persistent lipid metabolic imbalance causes lipotoxicity, "an abnormal fat storage in the peripheral organs". Hence, lipotoxicity could somewhat explain the process of sarcopenia, an aging-related, gradual, and involuntary decline in skeletal muscle strength and mass associated with several health complications. This review focuses on the recent mechanisms underlying lipotoxicity-related sarcopenia. A vicious cycle occurs between sarcopenia and ectopic fat storage via a complex interplay of mitochondrial dysfunction, pro-inflammatory cytokine production, oxidative stress, collagen deposition, extracellular matrix remodeling, and life habits. The repercussions of lipotoxicity exacerbation of sarcopenia can include increased disability, morbidity, and mortality. This suggests that appropriate lipotoxicity management should be considered the primary target for the prevention and/or treatment of chronic musculoskeletal and other aging-related disorders. Further advanced research is needed to understand the molecular details of lipotoxicity and its consequences for sarcopenia and sarcopenia-related comorbidities.
Collapse
Affiliation(s)
| | - Kamaluddin Hasan Motawei
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohammed Taha Al-Hariri
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia,Corresponding Author: Mohammed Taha Al-Hariri, Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia. E-mail:
| |
Collapse
|
7
|
Zhang J, Wen H, Qi X, Zhang Y, Dong X, Zhang K, Zhang M, Li J, Li Y. Morphological and Molecular Responses of Lateolabrax maculatus Skeletal Muscle Cells to Different Temperatures. Int J Mol Sci 2022; 23:ijms23179812. [PMID: 36077203 PMCID: PMC9456278 DOI: 10.3390/ijms23179812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/25/2022] Open
Abstract
Temperature strongly modulates muscle development and growth in ectothermic teleosts; however, the underlying mechanisms remain largely unknown. In this study, primary cultures of skeletal muscle cells of Lateolabrax maculatus were conducted and reared at different temperatures (21, 25, and 28 °C) in both the proliferation and differentiation stages. CCK-8, EdU, wound scratch and nuclear fusion index assays revealed that the proliferation, myogenic differentiation, and migration processes of skeletal muscle cells were significantly accelerated as the temperature raises. Based on the GO, GSEA, and WGCNA, higher temperature (28 °C) induced genes involved in HSF1 activation, DNA replication, and ECM organization processes at the proliferation stage, as well as HSF1 activation, calcium activity regulation, myogenic differentiation, and myoblast fusion, and sarcomere assembly processes at the differentiation stage. In contrast, lower temperature (21 °C) increased the expression levels of genes associated with DNA damage, DNA repair and apoptosis processes at the proliferation stage, and cytokine signaling and neutrophil degranulation processes at the differentiation stage. Additionally, we screened several hub genes regulating myogenesis processes. Our results could facilitate the understanding of the regulatory mechanism of temperature on fish skeletal muscle growth and further contribute to utilizing rational management strategies and promoting organism growth and development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yun Li
- Correspondence: ; Tel.: +86-0532-82-031-792
| |
Collapse
|
8
|
ITGβ6 Facilitates Skeletal Muscle Development by Maintaining the Properties and Cytoskeleton Stability of Satellite Cells. LIFE (BASEL, SWITZERLAND) 2022; 12:life12070926. [PMID: 35888016 PMCID: PMC9318838 DOI: 10.3390/life12070926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/11/2022] [Accepted: 06/16/2022] [Indexed: 11/25/2022]
Abstract
Integrin proteins are important receptors connecting the intracellular skeleton of satellite cells and the extracellular matrix (ECM), playing an important role in the process of skeletal muscle development. In this research, the function of ITGβ6 in regulating the differentiation of satellite cells was studied. Transcriptome and proteome analysis indicated that Itgβ6 is a key node connecting ECM-related proteins to the cytoskeleton, and it is necessary for the integrity of the membrane structure and stability of the cytoskeletal system, which are essential for satellite cell adhesion. Functional analysis revealed that the ITGβ6 protein could affect the myogenic differentiation potential of satellite cells by regulating the expression of PAX7 protein, thus regulating the formation of myotubes. Moreover, ITGβ6 is involved in muscle development by regulating cell-adhesion-related proteins, such as β-laminin, and cytoskeletal proteins such as PXN, DMD, and VCL. In conclusion, the effect of ITGβ6 on satellite cell differentiation mainly occurs before the initiation of differentiation, and it regulates terminal differentiation by affecting satellite cell characteristics, cell adhesion, and the stability of the cytoskeleton system.
Collapse
|
9
|
Orchard P, Manickam N, Ventresca C, Vadlamudi S, Varshney A, Rai V, Kaplan J, Lalancette C, Mohlke KL, Gallagher K, Burant CF, Parker SCJ. Human and rat skeletal muscle single-nuclei multi-omic integrative analyses nominate causal cell types, regulatory elements, and SNPs for complex traits. Genome Res 2021; 31:2258-2275. [PMID: 34815310 PMCID: PMC8647829 DOI: 10.1101/gr.268482.120] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/16/2021] [Indexed: 12/12/2022]
Abstract
Skeletal muscle accounts for the largest proportion of human body mass, on average, and is a key tissue in complex diseases and mobility. It is composed of several different cell and muscle fiber types. Here, we optimize single-nucleus ATAC-seq (snATAC-seq) to map skeletal muscle cell-specific chromatin accessibility landscapes in frozen human and rat samples, and single-nucleus RNA-seq (snRNA-seq) to map cell-specific transcriptomes in human. We additionally perform multi-omics profiling (gene expression and chromatin accessibility) on human and rat muscle samples. We capture type I and type II muscle fiber signatures, which are generally missed by existing single-cell RNA-seq methods. We perform cross-modality and cross-species integrative analyses on 33,862 nuclei and identify seven cell types ranging in abundance from 59.6% to 1.0% of all nuclei. We introduce a regression-based approach to infer cell types by comparing transcription start site-distal ATAC-seq peaks to reference enhancer maps and show consistency with RNA-based marker gene cell type assignments. We find heterogeneity in enrichment of genetic variants linked to complex phenotypes from the UK Biobank and diabetes genome-wide association studies in cell-specific ATAC-seq peaks, with the most striking enrichment patterns in muscle mesenchymal stem cells (∼3.5% of nuclei). Finally, we overlay these chromatin accessibility maps on GWAS data to nominate causal cell types, SNPs, transcription factor motifs, and target genes for type 2 diabetes signals. These chromatin accessibility profiles for human and rat skeletal muscle cell types are a useful resource for nominating causal GWAS SNPs and cell types.
Collapse
Affiliation(s)
- Peter Orchard
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Nandini Manickam
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Christa Ventresca
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Swarooparani Vadlamudi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Arushi Varshney
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Vivek Rai
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jeremy Kaplan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Claudia Lalancette
- Epigenomics Core, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Katherine Gallagher
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
10
|
Blackburn DM, Lazure F, Soleimani VD. SMART approaches for genome-wide analyses of skeletal muscle stem and niche cells. Crit Rev Biochem Mol Biol 2021; 56:284-300. [PMID: 33823731 DOI: 10.1080/10409238.2021.1908950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Darren M Blackburn
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
11
|
Extracellular matrix: an important regulator of cell functions and skeletal muscle development. Cell Biosci 2021; 11:65. [PMID: 33789727 PMCID: PMC8011170 DOI: 10.1186/s13578-021-00579-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Extracellular matrix (ECM) is a kind of connective tissue in the cell microenvironment, which is of great significance to tissue development. ECM in muscle fiber niche consists of three layers: the epimysium, the perimysium, and the endomysium (basal lamina). These three layers of connective tissue structure can not only maintain the morphology of skeletal muscle, but also play an important role in the physiological functions of muscle cells, such as the transmission of mechanical force, the regeneration of muscle fiber, and the formation of neuromuscular junction. In this paper, detailed discussions are made for the structure and key components of ECM in skeletal muscle tissue, the role of ECM in skeletal muscle development, and the application of ECM in biomedical engineering. This review will provide the reader with a comprehensive overview of ECM, as well as a comprehensive understanding of the structure, physiological function, and application of ECM in skeletal muscle tissue.
Collapse
|
12
|
Andreana I, Repellin M, Carton F, Kryza D, Briançon S, Chazaud B, Mounier R, Arpicco S, Malatesta M, Stella B, Lollo G. Nanomedicine for Gene Delivery and Drug Repurposing in the Treatment of Muscular Dystrophies. Pharmaceutics 2021; 13:278. [PMID: 33669654 PMCID: PMC7922331 DOI: 10.3390/pharmaceutics13020278] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/07/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Muscular Dystrophies (MDs) are a group of rare inherited genetic muscular pathologies encompassing a variety of clinical phenotypes, gene mutations and mechanisms of disease. MDs undergo progressive skeletal muscle degeneration causing severe health problems that lead to poor life quality, disability and premature death. There are no available therapies to counteract the causes of these diseases and conventional treatments are administered only to mitigate symptoms. Recent understanding on the pathogenetic mechanisms allowed the development of novel therapeutic strategies based on gene therapy, genome editing CRISPR/Cas9 and drug repurposing approaches. Despite the therapeutic potential of these treatments, once the actives are administered, their instability, susceptibility to degradation and toxicity limit their applications. In this frame, the design of delivery strategies based on nanomedicines holds great promise for MD treatments. This review focuses on nanomedicine approaches able to encapsulate therapeutic agents such as small chemical molecules and oligonucleotides to target the most common MDs such as Duchenne Muscular Dystrophy and the Myotonic Dystrophies. The challenge related to in vitro and in vivo testing of nanosystems in appropriate animal models is also addressed. Finally, the most promising nanomedicine-based strategies are highlighted and a critical view in future developments of nanomedicine for neuromuscular diseases is provided.
Collapse
Affiliation(s)
- Ilaria Andreana
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy;
| | - Mathieu Repellin
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (F.C.); (M.M.)
| | - Flavia Carton
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (F.C.); (M.M.)
- Department of Health Sciences, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy
| | - David Kryza
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
- Hospices Civils de Lyon, 69437 Lyon, France
| | - Stéphanie Briançon
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, University of Lyon, INSERM U1217, CNRS UMR 5310, 8 Avenue Rockefeller, 69008 Lyon, France; (B.C.); (R.M.)
| | - Rémi Mounier
- Institut NeuroMyoGène, University of Lyon, INSERM U1217, CNRS UMR 5310, 8 Avenue Rockefeller, 69008 Lyon, France; (B.C.); (R.M.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy;
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (F.C.); (M.M.)
| | - Barbara Stella
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy;
| | - Giovanna Lollo
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
| |
Collapse
|
13
|
Reid G, Magarotto F, Marsano A, Pozzobon M. Next Stage Approach to Tissue Engineering Skeletal Muscle. Bioengineering (Basel) 2020; 7:E118. [PMID: 33007935 PMCID: PMC7711907 DOI: 10.3390/bioengineering7040118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/18/2020] [Accepted: 09/26/2020] [Indexed: 02/08/2023] Open
Abstract
Large-scale muscle injury in humans initiates a complex regeneration process, as not only the muscular, but also the vascular and neuro-muscular compartments have to be repaired. Conventional therapeutic strategies often fall short of reaching the desired functional outcome, due to the inherent complexity of natural skeletal muscle. Tissue engineering offers a promising alternative treatment strategy, aiming to achieve an engineered tissue close to natural tissue composition and function, able to induce long-term, functional regeneration after in vivo implantation. This review aims to summarize the latest approaches of tissue engineering skeletal muscle, with specific attention toward fabrication, neuro-angiogenesis, multicellularity and the biochemical cues that adjuvate the regeneration process.
Collapse
Affiliation(s)
- Gregory Reid
- Department of Cardiac Surgery, University Hospital Basel, 4031 Basel, Switzerland; (G.R.); (A.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Fabio Magarotto
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Institute of Pediatric Research, Città della Speranza, 35127 Padova, Italy
| | - Anna Marsano
- Department of Cardiac Surgery, University Hospital Basel, 4031 Basel, Switzerland; (G.R.); (A.M.)
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Michela Pozzobon
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy;
- Institute of Pediatric Research, Città della Speranza, 35127 Padova, Italy
| |
Collapse
|
14
|
Trentesaux C, Striedinger K, Pomerantz JH, Klein OD. From gut to glutes: The critical role of niche signals in the maintenance and renewal of adult stem cells. Curr Opin Cell Biol 2020; 63:88-101. [PMID: 32036295 PMCID: PMC7247951 DOI: 10.1016/j.ceb.2020.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/17/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023]
Abstract
Stem cell behavior is tightly regulated by spatiotemporal signaling from the niche, which is a four-dimensional microenvironment that can instruct stem cells to remain quiescent, self-renew, proliferate, or differentiate. In this review, we discuss recent advances in understanding the signaling cues provided by the stem cell niche in two contrasting adult tissues, the rapidly cycling intestinal epithelium and the slowly renewing skeletal muscle. Drawing comparisons between these two systems, we discuss the effects of niche-derived growth factors and signaling molecules, metabolic cues, the extracellular matrix and biomechanical cues, and immune signals on stem cells. We also discuss the influence of the niche in defining stem cell identity and function in both normal and pathophysiologic states.
Collapse
Affiliation(s)
- Coralie Trentesaux
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Katharine Striedinger
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Jason H Pomerantz
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA; Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, USA.
| |
Collapse
|