1
|
Huot JR, Jamnick NA, Pin F, Livingston PD, Callaway CS, Bonetto A. GL261 glioblastoma induces delayed body weight gain and stunted skeletal muscle growth in young mice. Am J Physiol Regul Integr Comp Physiol 2025; 328:R628-R641. [PMID: 40247678 PMCID: PMC12123484 DOI: 10.1152/ajpregu.00035.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
The survival rate for children and adolescents has increased to over 85%. However, there is limited understanding of the impact of pediatric cancers on muscle development and physiology. Given that brain tumors alone account for 26% of all pediatric cancers, this study aimed to investigate the skeletal muscle consequences of tumor growth in young mice. C2C12 myotubes were cocultured with GL261 murine glioblastoma cells to assess myotube size. GL261 cells were then injected subcutaneously into 4-wk-old male C57BL/6J mice. Animals were euthanized 28 days post-GL261 implantation. Muscle function was tested in vivo and ex vivo. Muscle protein synthesis was estimated via the SUnSET method, and gene/protein expression levels were assessed via Western blotting and qPCR. In vitro, the C2C12 cultures exposed to GL261 exhibited myotube atrophy, consistent with a disrupted anabolic/catabolic balance. In vivo, carcass, heart, and fat mass were significantly reduced in the tumor-bearing mice. Skeletal muscle growth was impeded in the GL261 hosts, along with a smaller muscle cross-sectional area (CSA). Both in vivo muscle torque and the ex vivo Extensor Digitorum Longus (EDL) muscle force were unchanged. At molecular level, the tumor hosts displayed reduced estimations of muscle protein synthesis and increased muscle protein ubiquitination, in disagreement with decreased muscle ubiquitin ligase mRNA expression. Overall, we showed that GL261 tumors impact the growth of pediatric mice by stunting skeletal muscle development, decreasing muscle mass, reducing muscle fiber size, diminishing muscle protein synthesis, and altering protein catabolism signaling.NEW & NOTEWORTHY This study shows that pediatric brain tumors stunt muscle development in young mice. GL261 glioblastoma cells caused myotube atrophy, reduced carcass, heart, and fat mass, and impeded skeletal muscle growth. Tumor-bearing mice had decreased muscle protein synthesis and increased protein ubiquitination. This is the first demonstration that GL261 tumors reduce muscle mass and fiber size, impair muscle function and innervation, and alter muscle protein turnover.
Collapse
Affiliation(s)
- Joshua R. Huot
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | | | - Fabrizio Pin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Andrea Bonetto
- Department of Pathology
- Nutrition Obesity Research Center
- Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
2
|
Zhang X, Zhao M, Deng P, Liu H, Li H, Zhang X, Wang D, Qin J. Modeling Idiopathic Inflammatory Myopathy in the Bioinspired Muscle Tissue on Chip. Adv Healthc Mater 2025:e2405111. [PMID: 40357815 DOI: 10.1002/adhm.202405111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/21/2025] [Indexed: 05/15/2025]
Abstract
Idiopathic inflammatory myopathy (IIM) is an autoimmune disease that characterized by non-purulent inflammation of the skeletal muscle. However, due to the limitation of study model that can recapitulate the complex pathological process of IIM, the pathogenesis of IIM is still not fully clear. This manuscript develops a vascularized muscle tissue model on a chip that allows to model the immunity mediated pathological changes in IIM. This vascularized muscle model is constructed by layer-by-layer assembly, which could coculture of endothelial cells, myoblasts, and monocytes in a perfusable 3D system. The vascularized muscle model exhibits good biofunctions, including muscle cells alignment and fusion, myofibers generation, force production and expression of muscular biomarkers (myosin heavy chains 1, myosin heavy chains 7, actinin alpha 2, myogenin, and Desmin). Exposure to perfusion of activated monocytes, this work observes the functional changes of muscle tissue, which referred to myofibers atrophy, inflammatory response, and downregulated expression of muscle mature marker, consistent with clinical features of IIM. This work provides a unique platform for modelling IIM and paves a promising avenue for myopathies study and drug testing.
Collapse
Affiliation(s)
- Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Mengqian Zhao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengwei Deng
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Haitao Liu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Hongjing Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Xiaoqing Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Daqing Wang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
3
|
Oliver CE, Carter JL, Hong JS, Xu M, Kraus WE, Huffman KM, Truskey GA. Differential response of tissue engineered skeletal muscle from rheumatoid arthritis patients and healthy controls. Commun Biol 2025; 8:583. [PMID: 40200033 PMCID: PMC11978753 DOI: 10.1038/s42003-025-07970-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting articular joints and skeletal muscle. To assess the role of cytokines upon muscle strength in RA, we developed an in vitro tissue-engineered human skeletal muscle model (myobundle). Myobundles were generated using primary skeletal muscle cells from the vastus lateralis muscle of RA patients and age-matched healthy controls. RA myobundles were more sensitive to 5 ng/mL IFN-γ, exhibiting reduced contractile force and altered contraction kinetics. Addition of IL-6 with or without IFN-γ led to a small but significant increase in striated fibers. Gene sets involved in the response to hypoxia, MTOR1 signaling, and the unfolded protein response were enriched in IFN-γ-treated RA myobundles, but not IFN-γ-treated controls. Tofacitinib increased contractile force, myosin heavy chain, and PIM1 protein levels in RA myobundles treated with IFN-γ. Thus, in RA muscle, low levels of IFN-γ selectively increase gene pathways that reduce contractile force.
Collapse
Affiliation(s)
| | - Jonathan L Carter
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - James S Hong
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mingzhi Xu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - William E Kraus
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Kim M Huffman
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
4
|
Xiong G, Yu E, Heung M, Yang J, Lowe M, Abu-Hilal M. Weight gain secondary to the use of oral Janus kinase inhibitors: A systematic review and meta-analysis. JAAD Int 2025; 19:1-9. [PMID: 39872731 PMCID: PMC11763511 DOI: 10.1016/j.jdin.2024.11.009] [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] [Indexed: 01/30/2025] Open
Abstract
Oral Janus kinase inhibitors (JAKi) are increasingly used in dermatology, rheumatology, gastroenterology, and hematology. While effective, they can cause adverse effects such as acne, nausea, cytopenia, dyslipidemia, and Herpes zoster. Recent reports have linked JAKi usage to weight changes, particularly weight gain, which can significantly impact patients' quality of life. This study aimed to describe the incidence and characteristics of weight changes associated with the use of JAKi. Ovid MEDLINE, Embase, Web of Science, and Clinicaltrials.gov were searched up to April 2024. From 1080 initial articles, 90 studies covering 16,000 patients were selected. Our analysis found a notable incidence of weight gain with JAKi usage. Overall, 5.9% (947/16,000) of patients reported weight again. In randomized control trials, weight gain was observed in 7% (95% CI: 0.04; 0.09) of patients, while weight loss was observed in 1% (95% CI: 0.00; 0.03). Patients with dermatologic indications had lower weight gain rates (4%, 95% CI: 0.01; 0.06) than those with nondermatological indications (7%, 95% CI: 0.04; 0.10). Overall, JAKi therapy is associated with weight changes, particularly weight gain, underscoring the importance of appropriate counseling and weight monitoring. Further long-term studies are needed to better understand the mechanisms and management of JAKi-related weight changes.
Collapse
Affiliation(s)
- Grace Xiong
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Eric Yu
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Martin Heung
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Jaehyeong Yang
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Canada
| | - Megan Lowe
- Queen's University School of Medicine, Queens University, Kingston, Canada
| | | |
Collapse
|
5
|
Jia PW, Lin JZ, Zou YW, Ouyang ZM, Yang Y, Yang KM, Liang LH, Han JY, Yang ZH, Ma JD, Dai L. The Characteristic of Muscle Function for Sarcopenia in Patients with Rheumatoid Arthritis: A Large-Scale Real-World Cross-Sectional Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:551. [PMID: 40282842 PMCID: PMC12028369 DOI: 10.3390/medicina61040551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Sarcopenia is a notable comorbidity of rheumatoid arthritis (RA), affecting about one third of patients. However, the characteristic of muscle function and its association with RA disease remains unknown. Materials and Methods: This cross-sectional study collected clinical data from a real-world Chinese RA cohort. Sarcopenia was defined as both myopenia and low muscle function (LMF). Myopenia was defined as appendicular skeletal muscle mass index (ASMI) < 7.0 kg/m2 in men and <5.7 kg/m2 in women. LMF was defined as low muscle strength (LMS, hand grip < 28 kg in men and <18 kg in women) or low physical performance (LPP, 6 m gait speed < 1.0 m/s). Results: Among 1125 RA patients recruited in this study, 928 RA patients were eligible for analysis. The prevalence of sarcopenia, myopenia, LMF, LMS, and LPP in all RA patients was 36.5%, 46.1%, 69.0%, 57.8%, and 37.1%, respectively. According to their trends in age and disease activity, there were 111 (11.9%) patients in the young (age < 50 years) and remission (CDAI ≤ 2.8) subgroup, 199 (21.4%) patients in the young and active (CDAI > 2.8) subgroup, 198 (21.3%) patients in the old (age ≥ 50 years) and remission subgroup, and 420 (45.2%) patients in the old and active subgroup. Compared with the two remission subgroups, respectively, the young and active subgroup had significantly lower grip strength, higher prevalence of sarcopenia, LMF, and LMS, and worse activity function. After adjustment for potential confounders, multivariate multinominal logistic regression analysis showed that the young and active subgroup was positively associated with sarcopenia (OR = 3.193, 95%CI: 1.477-6.899), LMF (OR = 2.390, 95%CI: 1.207-4.731), and LMS (OR = 3.520, 95%CI: 1.743-7.110). Conclusions: Worse muscle strength, rather than reduced physical performance, is more common in patients with active RA at a young age. It underscores the critical need for early identification and intervention of muscle dysfunction to improve their quality of life.
Collapse
Affiliation(s)
- Pei-Wen Jia
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Jian-Zi Lin
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Yao-Wei Zou
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Zhi-Ming Ouyang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Ying Yang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Kui-Min Yang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Liu-Hong Liang
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Jin-Yuan Han
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Ze-Hong Yang
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China;
| | - Jian-Da Ma
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| | - Lie Dai
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou 510120, China; (P.-W.J.); (J.-Z.L.); (Y.-W.Z.); (Z.-M.O.); (Y.Y.); (K.-M.Y.); (L.-H.L.); (J.-Y.H.)
| |
Collapse
|
6
|
Huot JR, Jamnick NA, Pin F, Livingston PD, Callaway CS, Bonetto A. GL261 glioblastoma induces delayed body weight gain and stunted skeletal muscle growth in young mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.635159. [PMID: 39990490 PMCID: PMC11844426 DOI: 10.1101/2025.02.10.635159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Introduction The survival rate for children and adolescents has increased to over 85%. However, there is limited understanding of the impact of pediatric cancers on muscle development and physiology. Given that brain tumors alone account for 26% of all pediatric cancers, this study aimed to investigate the skeletal muscle consequences of tumor growth in young mice. Methods C2C12 myotubes were co-cultured with GL261 murine glioblastoma cells to assess myotube size. GL261 cells were then injected subcutaneously into 4-week-old male C57BL/6J mice. Animals were euthanized 28 days post-GL261 implantation. Muscle function was tested in vivo and ex vivo . Muscle protein synthesis was measured via the SUnSET method, and gene/protein expression levels were assessed via Western blotting and qPCR. Results In vitro , the C2C12 cultures exposed to GL261 exhibited myotube atrophy, consistent with a disrupted anabolic/catabolic balance. In vivo , carcass, heart, and fat mass were significantly reduced in the tumor-bearing mice. Skeletal muscle growth was impeded in the GL261 hosts, along with smaller muscle CSA. Both in vivo muscle torque and the ex vivo EDL muscle force were unchanged. At molecular level, the tumor hosts displayed reduced muscle protein synthesis and increased muscle protein ubiquitination, in disagreement with decreased muscle ubiquitin ligase mRNA expression. Conclusions Overall, we showed that GL261 tumors impact the growth of pediatric mice by stunting skeletal muscle development, decreasing muscle mass, reducing muscle fiber size, diminishing muscle protein synthesis, and altering protein catabolism signaling.
Collapse
|
7
|
DeLuca S, Strash N, Chen Y, Patsy M, Myers A, Tejeda L, Broders S, Miranda A, Jiang X, Bursac N. Engineered Cardiac Tissues as a Platform for CRISPR-Based Mitogen Discovery. Adv Healthc Mater 2025; 14:e2402201. [PMID: 39508305 PMCID: PMC11695184 DOI: 10.1002/adhm.202402201] [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: 06/15/2024] [Revised: 09/23/2024] [Indexed: 11/15/2024]
Abstract
Improved understanding of cardiomyocyte (CM) cell cycle regulation may allow researchers to stimulate pro-regenerative effects in injured hearts or promote maturation of human stem cell-derived CMs. Gene therapies, in particular, hold promise to induce controlled proliferation of endogenous or transplanted CMs via transient activation of mitogenic processes. Methods to identify and characterize candidate cardiac mitogens in vitro can accelerate translational efforts and contribute to the understanding of the complex regulatory landscape of CM proliferation and postnatal maturation. In this study, A CRISPR knockout-based screening strategy using in vitro neonatal rat ventricular myocyte (NRVM) monolayers is established, followed by candidate mitogen validation in mature 3-D engineered cardiac tissues (ECTs). This screen identified knockout of the purine metabolism enzyme adenosine deaminase (ADA-KO) as an effective pro-mitogenic stimulus. RNA-sequencing of ECTs further reveals increased pentose phosphate pathway (PPP) activity as the primary driver of ADA-KO-induced CM cycling. Inhibition of the pathway's rate limiting enzyme, glucose-6-phosphate dehydrogenase (G6PD), prevented ADA-KO induced CM cycling, while increasing PPP activity via G6PD overexpression increased CM cycling. Together, this study demonstrates the development and application of a genetic/tissue engineering platform for in vitro discovery and validation of new candidate mitogens affecting regenerative or maturation states of cardiomyocytes.
Collapse
Affiliation(s)
- Sophia DeLuca
- Department of Biomedical Engineering
- Department of Cell Biology, Duke University, Durham, NC, 27708, USA
| | - Nicholas Strash
- Department of Biomedical Engineering
- Department of Cell Biology, Duke University, Durham, NC, 27708, USA
| | | | | | | | | | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering
- Department of Cell Biology, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
8
|
Zhang C, Jing Y, Wang J, Xia Z, Lai Y, Bai L, Su J. Skeletal organoids. BIOMATERIALS TRANSLATIONAL 2024; 5:390-410. [PMID: 39872931 PMCID: PMC11764188 DOI: 10.12336/biomatertransl.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 01/30/2025]
Abstract
The skeletal system, composed of bones, muscles, joints, ligaments, and tendons, serves as the foundation for maintaining human posture, mobility, and overall biomechanical functionality. However, with ageing, chronic overuse, and acute injuries, conditions such as osteoarthritis, intervertebral disc degeneration, muscle atrophy, and ligament or tendon tears have become increasingly prevalent and pose serious clinical challenges. These disorders not only result in pain, functional loss, and a marked reduction in patients' quality of life but also impose substantial social and economic burdens. Current treatment modalities, including surgical intervention, pharmacotherapy, and physical rehabilitation, often do not effectively restore the functionality of damaged tissues and are associated with high recurrence rates and long-term complications, highlighting significant limitations in their efficacy. Thus, there is a strong demand to develop novel and more effective therapeutic and reparative strategies. Organoid technology, as a three-dimensional micro-tissue model, can replicate the structural and functional properties of native tissues in vitro, providing a novel platform for in-depth studies of disease mechanisms, optimisation of drug screening, and promotion of tissue regeneration. In recent years, substantial advancements have been made in the research of bone, muscle, and joint organoids, demonstrating their broad application potential in personalised and regenerative medicine. Nonetheless, a comprehensive review of current research on skeletal organoids is still lacking. Therefore, this article aims to present an overview of the definition and technological foundation of organoids, systematically summarise the progress in the construction and application of skeletal organoids, and explore future opportunities and challenges in this field, offering valuable insights and references for researchers.
Collapse
Affiliation(s)
- Chen Zhang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yingying Jing
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Jianhua Wang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhidao Xia
- Centre for Nanohealth, Swansea University Medical School, Swansea University, Swansea, UK
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang Province, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Broer T, Tsintolas N, Purkey K, Hammond S, DeLuca S, Wu T, Gupta I, Khodabukus A, Bursac N. Engineered myovascular tissues for studies of endothelial/satellite cell interactions. Acta Biomater 2024; 188:65-78. [PMID: 39299621 PMCID: PMC11486565 DOI: 10.1016/j.actbio.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
In native skeletal muscle, capillaries reside in close proximity to muscle stem cells (satellite cells, SCs) and regulate SC numbers and quiescence through partially understood mechanisms that are difficult to study in vivo. This challenge could be addressed by the development of a 3-dimensional (3D) in vitro model of vascularized skeletal muscle harboring both a pool of quiescent SCs and a robust network of capillaries. Still, studying interactions between SCs and endothelial cells (ECs) within a tissue-engineered muscle environment has been hampered by the incompatibility of commercially available EC media with skeletal muscle differentiation. In this study, we first optimized co-culture media and cellular ratios to generate highly functional vascularized human skeletal muscle tissues ("myovascular bundles") with contractile properties (∼10 mN/mm2) equaling those of avascular, muscle-only tissues ("myobundles"). Within one week of muscle differentiation, ECs in these tissues formed a dense network of capillaries that co-aligned with muscle fibers and underwent initial lumenization. Incorporating vasculature within myobundles increased the total SC number by 82%, with SC density and quiescent signature being increased proximal (≤20μm) to EC networks. In vivo, at two weeks post-implantation into dorsal window chambers in nude mice, vascularized myobundles exhibited improved calcium handling compared to avascular implants. In summary, we engineered highly functional myovascular tissues that enable studies of the roles of EC-SC crosstalk in human muscle development, physiology, and disease. STATEMENT OF SIGNIFICANCE: In native skeletal muscle, intricate relationships between vascular cells and muscle stem cells ("satellite cells") play critical roles in muscle growth and regeneration. Current methods for in vitro engineering of contractile skeletal muscle do not recreate capillary networks present in vivo. Our study for the first time generates in vitro robustly vascularized, highly functional engineered human skeletal muscle tissues. Within these tissues, satellite cells are more abundant and, similar to in vivo, they are more dense and less proliferative proximal to endothelial cells. Upon implantation in mice, vascularized engineered muscles show improved calcium handling compared to muscle-only implants. We expect that this versatile in vitro system will enable studies of muscle-vasculature crosstalk in human development and disease.
Collapse
Affiliation(s)
- Torie Broer
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Nick Tsintolas
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Karly Purkey
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Stewart Hammond
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Sophia DeLuca
- Department of Cell Biology, Duke University, Durham, NC 27708, USA
| | - Tianyu Wu
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Ishika Gupta
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
10
|
Covert LT, Prinz JA, Swain-Lenz D, Dvergsten J, Truskey GA. Genetic changes from type I interferons and JAK inhibitors: clues to drivers of juvenile dermatomyositis. Rheumatology (Oxford) 2024; 63:SI240-SI248. [PMID: 38317053 PMCID: PMC11381683 DOI: 10.1093/rheumatology/keae082] [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: 11/09/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE To better understand the pathogenesis of juvenile dermatomyositis (JDM), we examined the effect of the cytokines type I interferons (IFN I) and JAK inhibitor drugs (JAKi) on gene expression in bioengineered pediatric skeletal muscle. METHODS Myoblasts from three healthy pediatric donors were used to create three-dimensional skeletal muscle units termed myobundles. Myobundles were treated with IFN I, either IFNα or IFNβ. A subset of IFNβ-exposed myobundles was treated with JAKi tofacitinib or baricitinib. RNA sequencing analysis was performed on all myobundles. RESULTS Seventy-six myobundles were analysed. Principal component analysis showed donor-specific clusters of gene expression across IFNα and IFNβ-exposed myobundles in a dose-dependent manner. Both cytokines upregulated interferon response and proinflammatory genes; however, IFNβ led to more significant upregulation. Key downregulated pathways involved oxidative phosphorylation, fatty acid metabolism and myogenesis genes. Addition of tofacitinib or baricitinib moderated the gene expression induced by IFNβ, with partial reversal of upregulated inflammatory and downregulated myogenesis pathways. Baricitinib altered genetic profiles more than tofacitinib. CONCLUSION IFNβ leads to more pro-inflammatory gene upregulation than IFNα, correlating to greater decrease in contractile protein gene expression and reduced contractile force. JAK inhibitors, baricitinib more so than tofacitinib, partially reverse IFN I-induced genetic changes. Increased IFN I exposure in healthy bioengineered skeletal muscle leads to IFN-inducible gene expression, inflammatory pathway enrichment, and myogenesis gene downregulation, consistent with what is observed in JDM.
Collapse
Affiliation(s)
- Lauren T Covert
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - Joseph A Prinz
- Sequencing and Genomics Technologies Core Facility, School of Medicine, Duke University, Durham, NC, USA
| | - Devjanee Swain-Lenz
- Sequencing and Genomics Technologies Core Facility, School of Medicine, Duke University, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Jeffrey Dvergsten
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
11
|
Huang Y, Yao K, Zhang Q, Huang X, Chen Z, Zhou Y, Yu X. Bioelectronics for electrical stimulation: materials, devices and biomedical applications. Chem Soc Rev 2024; 53:8632-8712. [PMID: 39132912 DOI: 10.1039/d4cs00413b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Bioelectronics is a hot research topic, yet an important tool, as it facilitates the creation of advanced medical devices that interact with biological systems to effectively diagnose, monitor and treat a broad spectrum of health conditions. Electrical stimulation (ES) is a pivotal technique in bioelectronics, offering a precise, non-pharmacological means to modulate and control biological processes across molecular, cellular, tissue, and organ levels. This method holds the potential to restore or enhance physiological functions compromised by diseases or injuries by integrating sophisticated electrical signals, device interfaces, and designs tailored to specific biological mechanisms. This review explains the mechanisms by which ES influences cellular behaviors, introduces the essential stimulation principles, discusses the performance requirements for optimal ES systems, and highlights the representative applications. From this review, we can realize the potential of ES based bioelectronics in therapy, regenerative medicine and rehabilitation engineering technologies, ranging from tissue engineering to neurological technologies, and the modulation of cardiovascular and cognitive functions. This review underscores the versatility of ES in various biomedical contexts and emphasizes the need to adapt to complex biological and clinical landscapes it addresses.
Collapse
Affiliation(s)
- Ya Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kuanming Yao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Qiang Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xingcan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Zhenlin Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yu Zhou
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
12
|
Bakinowska E, Olejnik-Wojciechowska J, Kiełbowski K, Skoryk A, Pawlik A. Pathogenesis of Sarcopenia in Chronic Kidney Disease-The Role of Inflammation, Metabolic Dysregulation, Gut Dysbiosis, and microRNA. Int J Mol Sci 2024; 25:8474. [PMID: 39126043 PMCID: PMC11313360 DOI: 10.3390/ijms25158474] [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: 05/31/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Chronic kidney disease (CKD) is a progressive disorder associated with a decline in kidney function. Consequently, patients with advanced stages of CKD require renal replacement therapies, such as dialysis and kidney transplantation. Various conditions lead to the development of CKD, including diabetes mellitus, hypertension, and glomerulonephritis, among others. The disease is associated with metabolic and hormonal dysregulation, including uraemia and hyperparathyroidism, as well as with low-grade systemic inflammation. Altered homeostasis increases the risk of developing severe comorbidities, such as cardiovascular diseases or sarcopenia, which increase mortality. Sarcopenia is defined as a progressive decline in muscle mass and function. However, the precise mechanisms that link CKD and the development of sarcopenia are poorly understood. Knowledge about these linking mechanisms might lead to the introduction of precise treatment strategies that could prevent muscle wasting. This review discusses inflammatory mediators, metabolic and hormonal dysregulation, gut microbiota dysbiosis, and non-coding RNA alterations that could link CKD and sarcopenia.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| | - Joanna Olejnik-Wojciechowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
- Independent Laboratory of Community Nursing, Pomeranian Medical University, 71-210 Szczecin, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| | - Anastasiia Skoryk
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (E.B.); (J.O.-W.); (K.K.); (A.S.)
| |
Collapse
|
13
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
14
|
Kim JW, Bae JH, Go GY, Lee JR, Jeong Y, Kim JY, Kim TH, Kim YK, Han JW, Oh JE, Hahn MJ, Kang JS, Bae GU. Epsti1 Regulates the Inflammatory Stage of Early Muscle Regeneration through STAT1-VCP Interaction. Int J Biol Sci 2024; 20:3530-3543. [PMID: 38993551 PMCID: PMC11234217 DOI: 10.7150/ijbs.94675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
During muscle regeneration, interferon-gamma (IFN-γ) coordinates inflammatory responses critical for activation of quiescent muscle stem cells upon injury via the Janus kinase (JAK) - signal transducer and activator of transcription 1 (STAT1) pathway. Dysregulation of JAK-STAT1 signaling results in impaired muscle regeneration, leading to muscle dysfunction or muscle atrophy. Until now, the underlying molecular mechanism of how JAK-STAT1 signaling resolves during muscle regeneration remains largely elusive. Here, we demonstrate that epithelial-stromal interaction 1 (Epsti1), an interferon response gene, has a crucial role in regulating the IFN-γ-JAK-STAT1 signaling at early stage of muscle regeneration. Epsti1-deficient mice exhibit impaired muscle regeneration with elevated inflammation response. In addition, Epsti1-deficient myoblasts display aberrant interferon responses. Epsti1 interacts with valosin-containing protein (VCP) and mediates the proteasomal degradation of IFN-γ-activated STAT1, likely contributing to dampening STAT1-mediated inflammation. In line with the notion, mice lacking Epsti1 exhibit exacerbated muscle atrophy accompanied by increased inflammatory response in cancer cachexia model. Our study suggests a crucial function of Epsti1 in the resolution of IFN-γ-JAK-STAT1 signaling through interaction with VCP which provides insights into the unexplored mechanism of crosstalk between inflammatory response and muscle regeneration.
Collapse
Affiliation(s)
- Jee Won Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Ju-Hyeon Bae
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
| | - Ga-Yeon Go
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jae-Rin Lee
- Cell and Gene Therapy Products Division, National Institute of Food and Drug Safety Evaluation, Cheongju 28159, South Korea
| | - Yideul Jeong
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| | - Jun-Young Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Tae Hyun Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Yong Kee Kim
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
| | - Jeung-Whan Han
- Research Center for Epigenome Regulation, School of Pharmacy, Department of Biochemistry and Molecular Biology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Ji-Eun Oh
- Department of Biomedical Laboratory Science, Far East University, 76-32 Daehakgil, Gamgok-myeon, Eumseong-gun, Chungbuk-do, 27601, Korea
| | - Myong-Joon Hahn
- Research Center for Epigenome Regulation, School of Pharmacy, Department of Biochemistry and Molecular Biology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| | - Gyu-Un Bae
- Drug Information Research Institute, Sookmyung Women's University, Seoul 04310, South Korea
- Muscle Physiome Research Center, Sookmyung Women's University, Seoul 04310, South Korea
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, South Korea
- Research Institute of Aging Related Disease, AniMusCure Inc., Suwon 16419, South Korea
| |
Collapse
|
15
|
Bombieri C, Corsi A, Trabetti E, Ruggiero A, Marchetto G, Vattemi G, Valenti MT, Zipeto D, Romanelli MG. Advanced Cellular Models for Rare Disease Study: Exploring Neural, Muscle and Skeletal Organoids. Int J Mol Sci 2024; 25:1014. [PMID: 38256087 PMCID: PMC10815694 DOI: 10.3390/ijms25021014] [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: 12/06/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Organoids are self-organized, three-dimensional structures derived from stem cells that can mimic the structure and physiology of human organs. Patient-specific induced pluripotent stem cells (iPSCs) and 3D organoid model systems allow cells to be analyzed in a controlled environment to simulate the characteristics of a given disease by modeling the underlying pathophysiology. The recent development of 3D cell models has offered the scientific community an exceptionally valuable tool in the study of rare diseases, overcoming the limited availability of biological samples and the limitations of animal models. This review provides an overview of iPSC models and genetic engineering techniques used to develop organoids. In particular, some of the models applied to the study of rare neuronal, muscular and skeletal diseases are described. Furthermore, the limitations and potential of developing new therapeutic approaches are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (C.B.); (A.C.); (E.T.); (A.R.); (G.M.); (G.V.); (M.T.V.)
| | - Maria Grazia Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (C.B.); (A.C.); (E.T.); (A.R.); (G.M.); (G.V.); (M.T.V.)
| |
Collapse
|
16
|
Covert LT, Patel H, Osman A, Duncan L, Dvergsten J, Truskey GA. Effect of type I interferon on engineered pediatric skeletal muscle: a promising model for juvenile dermatomyositis. Rheumatology (Oxford) 2024; 63:209-217. [PMID: 37094222 PMCID: PMC10765138 DOI: 10.1093/rheumatology/kead186] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023] Open
Abstract
OBJECTIVE To investigate pathogenic mechanisms underlying JDM, we defined the effect of type I IFN, IFN-α and IFN-β, on pediatric skeletal muscle function and expression of myositis-related proteins using an in vitro engineered human skeletal muscle model (myobundle). METHODS Primary myoblasts were isolated from three healthy pediatric donors and used to create myobundles that mimic functioning skeletal muscle in structural architecture and physiologic function. Myobundles were exposed to 0, 5, 10 or 20 ng/ml IFN-α or IFN-β for 7 days and then functionally tested under electrical stimulation and analyzed immunohistochemically for structural and myositis-related proteins. Additionally, IFN-β-exposed myobundles were treated with Janus kinase inhibitors (JAKis) tofacitinib and baricitinib. These myobundles were also analyzed for contractile force and immunohistochemistry. RESULTS IFN-β, but not IFN-α, was associated with decreased contractile tetanus force and slowed twitch kinetics. These effects were reversed by tofacitinib and baricitinib. Type I IFN paradoxically reduced myobundle fatigue, which did not reverse after JAKi. Additionally, type I IFN correlated with MHC I upregulation, which normalized after JAKi treatment, but expression of myositis-specific autoantigens Mi-2, melanocyte differentiation-associated protein 5 and the endoplasmic reticulum stress marker GRP78 were variable and donor specific after type I IFN exposure. CONCLUSION IFN-α and IFN-β have distinct effects on pediatric skeletal muscle and these effects can partially be reversed by JAKi treatment. This is the first study illustrating effective use of a three-dimensional human skeletal muscle model to investigate JDM pathogenesis and test novel therapeutics.
Collapse
Affiliation(s)
- Lauren T Covert
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Alaa Osman
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Lavonia Duncan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jeffrey Dvergsten
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
17
|
Assyov Y, Ganeva I, Ikonomov S, Nedeva I, Velikov T, Kamenov Z, Velikova T. Interleukin-6: Unravelling its role in sarcopenia pathogenesis and exploring therapeutic avenues. PHARMACIA 2023; 70:1493-1498. [DOI: 10.3897/pharmacia.70.e115762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
This review explores the intricate relationship between interleukin-6 (IL-6) and sarcopenia, a prevalent condition characterized by progressive skeletal muscle loss, particularly in aging populations. Emphasizing the rising prevalence and health challenges posed by sarcopenia, the paper delves into the multifunctional roles of IL-6 in immune response, inflammation and inflammaging associated with sarcopenia. Significantly elevated in sarcopenic individuals, IL-6 prompts an exploration of its molecular impact on muscle wasting. The review critically assesses IL-6 as a potential biomarker for sarcopenia diagnosis and prognosis while also examining therapeutic interventions targeting IL-6 signaling pathways, offering a foundation for future research and the development of targeted therapeutic strategies to alleviate the impact of this debilitating condition.
Collapse
|
18
|
Rizk J, Sahu R, Duteil D. An overview on androgen-mediated actions in skeletal muscle and adipose tissue. Steroids 2023; 199:109306. [PMID: 37634653 DOI: 10.1016/j.steroids.2023.109306] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Androgens are a class of steroid hormones primarily associated with male sexual development and physiology, but exert pleiotropic effects in either sex. They have a crucial role in various physiological processes, including the regulation of skeletal muscle and adipose tissue homeostasis. The effects of androgens are mainly mediated through the androgen receptor (AR), a ligand-activated nuclear receptor expressed in both tissues. In skeletal muscle, androgens via AR exert a multitude of effects, ranging from increased muscle mass and strength, to the regulation of muscle fiber type composition, contraction and metabolic functions. In adipose tissue, androgens influence several processes including proliferation, fat distribution, and metabolism but they display depot-specific and organism-specific effects which differ in certain context. This review further explores the potential mechanisms underlying androgen-AR signaling in skeletal muscle and adipose tissue. Understanding the roles of androgens and their receptor in skeletal muscle and adipose tissue is essential for elucidating their contributions to physiological processes, disease conditions, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Joe Rizk
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Rajesh Sahu
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France
| | - Delphine Duteil
- Université de Strasbourg, CNRS, Inserm, IGBMC UMR 7104- UMR-S 1258, F-67400 Illkirch, France.
| |
Collapse
|
19
|
Wang K, Smith SH, Iijima H, Hettinger ZR, Mallepally A, Shroff SG, Ambrosio F. Bioengineered 3D Skeletal Muscle Model Reveals Complement 4b as a Cell-Autonomous Mechanism of Impaired Regeneration with Aging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207443. [PMID: 36650030 DOI: 10.1002/adma.202207443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/11/2022] [Indexed: 05/17/2023]
Abstract
A mechanistic understanding of cell-autonomous skeletal muscle changes after injury can lead to novel interventions to improve functional recovery in an aged population. However, major knowledge gaps persist owing to limitations of traditional biological aging models. 2D cell culture represents an artificial environment, while aging mammalian models are contaminated by influences from non-muscle cells and other organs. Here, a 3D muscle aging system is created to overcome the limitations of these traditional platforms. It is shown that old muscle constructs (OMC) manifest a sarcopenic phenotype, as evidenced by hypotrophic myotubes, reduced contractile function, and decreased regenerative capacity compared to young muscle constructs. OMC also phenocopy the regenerative responses of aged muscle to two interventions, pharmacological and biological. Interrogation of muscle cell-specific mechanisms that contribute to impaired regeneration over time further reveals that an aging-induced increase of complement component 4b (C4b) delays muscle progenitor cell amplification and impairs functional recovery. However, administration of complement factor I, a C4b inactivator, improves muscle regeneration in vitro and in vivo, indicating that C4b inhibition may be a novel approach to enhance aged muscle repair. Collectively, the model herein exhibits capabilities to study cell-autonomous changes in skeletal muscle during aging, regeneration, and intervention.
Collapse
Affiliation(s)
- Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephen H Smith
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Zachary R Hettinger
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Medicine, Division of Geriatric Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Adarsh Mallepally
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sanjeev G Shroff
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Charlestown, MA, 02129, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, MA, 02115, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
20
|
Bennett JL, Pratt AG, Dodds R, Sayer AA, Isaacs JD. Rheumatoid sarcopenia: loss of skeletal muscle strength and mass in rheumatoid arthritis. Nat Rev Rheumatol 2023; 19:239-251. [PMID: 36801919 DOI: 10.1038/s41584-023-00921-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2023] [Indexed: 02/19/2023]
Abstract
Sarcopenia, a disorder that involves the generalized loss of skeletal muscle strength and mass, was formally recognized as a disease by its inclusion in the International Classification of Diseases in 2016. Sarcopenia typically affects older people, but younger individuals with chronic disease are also at risk. The risk of sarcopenia is high (with a prevalence of ≥25%) in individuals with rheumatoid arthritis (RA), and this rheumatoid sarcopenia is associated with increased likelihood of falls, fractures and physical disability, in addition to the burden of joint inflammation and damage. Chronic inflammation mediated by cytokines such as TNF, IL-6 and IFNγ contributes to aberrant muscle homeostasis (for instance, by exacerbating muscle protein breakdown), and results from transcriptomic studies have identified dysfunction of muscle stem cells and metabolism in RA. Progressive resistance exercise is an effective therapy for rheumatoid sarcopenia but it can be challenging or unsuitable for some individuals. The unmet need for anti-sarcopenia pharmacotherapies is great, both for people with RA and for otherwise healthy older adults.
Collapse
Affiliation(s)
- Joshua L Bennett
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK.
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Richard Dodds
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Avan A Sayer
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
21
|
McNamara SL, Seo BR, Freedman BR, Roloson EB, Alvarez JT, O'Neill CT, Vandenburgh HH, Walsh CJ, Mooney DJ. Anti-inflammatory therapy enables robot-actuated regeneration of aged muscle. Sci Robot 2023; 8:eadd9369. [PMID: 36947599 DOI: 10.1126/scirobotics.add9369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Robot-actuated mechanical loading (ML)-based therapies ("mechanotherapies") can promote regeneration after severe skeletal muscle injury, but the effectiveness of such approaches during aging is unknown and may be influenced by age-associated decline in the healing capacity of skeletal muscle. To address this knowledge gap, this work used a noninvasive, load-controlled robotic device to impose highly defined tissue stresses to evaluate the age dependence of ML on muscle repair after injury. The response of injured muscle to robot-actuated cyclic compressive loading was found to be age sensitive, revealing not only a lack of reparative benefit of ML on injured aged muscles but also exacerbation of tissue inflammation. ML alone also disrupted the normal regenerative processes of aged muscle stem cells. However, these negative effects could be reversed by introducing anti-inflammatory therapy alongside ML application, leading to enhanced skeletal muscle regeneration even in aged mice.
Collapse
Affiliation(s)
- S L McNamara
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - B R Seo
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - B R Freedman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - E B Roloson
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - J T Alvarez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - C T O'Neill
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - H H Vandenburgh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - C J Walsh
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - D J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| |
Collapse
|
22
|
Venturini G, Alvim JM, Padilha K, Toepfer CN, Gorham JM, Wasson LK, Biagi D, Schenkman S, Carvalho VM, Salgueiro JS, Cardozo KHM, Krieger JE, Pereira AC, Seidman JG, Seidman CE. Cardiomyocyte infection by Trypanosoma cruzi promotes innate immune response and glycolysis activation. Front Cell Infect Microbiol 2023; 13:1098457. [PMID: 36814444 PMCID: PMC9940271 DOI: 10.3389/fcimb.2023.1098457] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023] Open
Abstract
Introduction Chagas cardiomyopathy, a disease caused by Trypanosoma cruzi (T. cruzi) infection, is a major contributor to heart failure in Latin America. There are significant gaps in our understanding of the mechanism for infection of human cardiomyocytes, the pathways activated during the acute phase of the disease, and the molecular changes that lead to the progression of cardiomyopathy. Methods To investigate the effects of T. cruzi on human cardiomyocytes during infection, we infected induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) with the parasite and analyzed cellular, molecular, and metabolic responses at 3 hours, 24 hours, and 48 hours post infection (hpi) using transcriptomics (RNAseq), proteomics (LC-MS), and metabolomics (GC-MS and Seahorse) analyses. Results Analyses of multiomic data revealed that cardiomyocyte infection caused a rapid increase in genes and proteins related to activation innate and adaptive immune systems and pathways, including alpha and gamma interferons, HIF-1α signaling, and glycolysis. These responses resemble prototypic responses observed in pathogen-activated immune cells. Infection also caused an activation of glycolysis that was dependent on HIF-1α signaling. Using gene editing and pharmacological inhibitors, we found that T. cruzi uptake was mediated in part by the glucose-facilitated transporter GLUT4 and that the attenuation of glycolysis, HIF-1α activation, or GLUT4 expression decreased T. cruzi infection. In contrast, pre-activation of pro-inflammatory immune responses with LPS resulted in increased infection rates. Conclusion These findings suggest that T. cruzi exploits a HIF-1α-dependent, cardiomyocyte-intrinsic stress-response activation of glycolysis to promote intracellular infection and replication. These chronic immuno-metabolic responses by cardiomyocytes promote dysfunction, cell death, and the emergence of cardiomyopathy.
Collapse
Affiliation(s)
- Gabriela Venturini
- Department of Genetics, Harvard Medical School, Boston, MA, United States,Laboratory of Genetics and Molecular Cardiology, University of São Paulo Medical School, São Paulo, Brazil
| | - Juliana M. Alvim
- Laboratory of Genetics and Molecular Cardiology, University of São Paulo Medical School, São Paulo, Brazil
| | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, University of São Paulo Medical School, São Paulo, Brazil
| | - Christopher N. Toepfer
- Department of Genetics, Harvard Medical School, Boston, MA, United States,Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom,Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Joshua M. Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Lauren K. Wasson
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | | | - Sergio Schenkman
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, São Paulo, Brazil
| | | | | | | | - Jose E. Krieger
- Laboratory of Genetics and Molecular Cardiology, University of São Paulo Medical School, São Paulo, Brazil
| | - Alexandre C. Pereira
- Department of Genetics, Harvard Medical School, Boston, MA, United States,Laboratory of Genetics and Molecular Cardiology, University of São Paulo Medical School, São Paulo, Brazil
| | | | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, United States,Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States,Howard Hughes Medical Institute, Chevy Chase, MD, United States,*Correspondence: Christine E. Seidman,
| |
Collapse
|
23
|
Ostrovidov S, Ramalingam M, Bae H, Orive G, Fujie T, Shi X, Kaji H. Latest developments in engineered skeletal muscle tissues for drug discovery and development. Expert Opin Drug Discov 2023; 18:47-63. [PMID: 36535280 DOI: 10.1080/17460441.2023.2160438] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION With the advances in skeletal muscle tissue engineering, new platforms have arisen with important applications in biology studies, disease modeling, and drug testing. Current developments highlight the quest for engineering skeletal muscle tissues with higher complexity . These new human skeletal muscle tissue models will be powerful tools for drug discovery and development and disease modeling. AREAS COVERED The authors review the latest advances in in vitro models of engineered skeletal muscle tissues used for testing drugs with a focus on the use of four main cell culture techniques: Cell cultures in well plates, in microfluidics, in organoids, and in bioprinted constructs. Additional information is provided on the satellite cell niche. EXPERT OPINION In recent years, more sophisticated in vitro models of skeletal muscle tissues have been fabricated. Important developments have been made in stem cell research and in the engineering of human skeletal muscle tissue. Some platforms have already started to be used for drug testing, notably those based on the parameters of hypertrophy/atrophy and the contractibility of myotubes. More developments are expected through the use of multicellular types and multi-materials as matrices . The validation and use of these models in drug testing should now increase.
Collapse
Affiliation(s)
- Serge Ostrovidov
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science, BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,School of Basic Medical Science, Chengdu University, Chengdu, Sichuan, China.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,Department of Metallurgical and Materials Engineering, Atilim University, Ankara, Turkey
| | - Hojae Bae
- KU Convergence Science and Technology Institute, Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.,Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.,Biomaterials and Nanomedicine (CIBER-BBN), Biomedical Research Networking Centre in Bioengineering, Vitoria-Gasteiz, Spain
| | - Toshinori Fujie
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
24
|
Laurila PP, Wohlwend M, Imamura de Lima T, Luan P, Herzig S, Zanou N, Crisol B, Bou-Sleiman M, Porcu E, Gallart-Ayala H, Handzlik MK, Wang Q, Jain S, D'Amico D, Salonen M, Metallo CM, Kutalik Z, Eichmann TO, Place N, Ivanisevic J, Lahti J, Eriksson JG, Auwerx J. Sphingolipids accumulate in aged muscle, and their reduction counteracts sarcopenia. NATURE AGING 2022; 2:1159-1175. [PMID: 37118545 DOI: 10.1038/s43587-022-00309-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/07/2022] [Indexed: 04/30/2023]
Abstract
Age-related muscle dysfunction and sarcopenia are major causes of physical incapacitation in older adults and currently lack viable treatment strategies. Here we find that sphingolipids accumulate in mouse skeletal muscle upon aging and that both genetic and pharmacological inhibition of sphingolipid synthesis prevent age-related decline in muscle mass while enhancing strength and exercise capacity. Inhibition of sphingolipid synthesis confers increased myogenic potential and promotes protein synthesis. Within the sphingolipid pathway, we show that accumulation of dihydroceramides is the culprit disturbing myofibrillar homeostasis. The relevance of sphingolipid pathways in human aging is demonstrated in two cohorts, the UK Biobank and Helsinki Birth Cohort Study in which gene expression-reducing variants of SPTLC1 and DEGS1 are associated with improved and reduced fitness of older individuals, respectively. These findings identify sphingolipid synthesis inhibition as an attractive therapeutic strategy for age-related sarcopenia and co-occurring pathologies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sébastien Herzig
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maroun Bou-Sleiman
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eleonora Porcu
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Michal K Handzlik
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Qi Wang
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Suresh Jain
- Intonation Research Laboratories, Secunderabad, India
| | - Davide D'Amico
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Minna Salonen
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Christian M Metallo
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zoltan Kutalik
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Center for Explorative Lipidomics, BioTechMed-Graz, Graz, Austria
| | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, University of Helsinki, Helsinki, Finland
- Department of Obstetrics and Gynecology, National University Singapore, Yong Loo Lin School of Medicine, Singapore, Singapore
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
25
|
Sanchez MM, Bagdasarian IA, Darch W, Morgan JT. Organotypic cultures as aging associated disease models. Aging (Albany NY) 2022; 14:9338-9383. [PMID: 36435511 PMCID: PMC9740367 DOI: 10.18632/aging.204361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/21/2022] [Indexed: 11/24/2022]
Abstract
Aging remains a primary risk factor for a host of diseases, including leading causes of death. Aging and associated diseases are inherently multifactorial, with numerous contributing factors and phenotypes at the molecular, cellular, tissue, and organismal scales. Despite the complexity of aging phenomena, models currently used in aging research possess limitations. Frequently used in vivo models often have important physiological differences, age at different rates, or are genetically engineered to match late disease phenotypes rather than early causes. Conversely, routinely used in vitro models lack the complex tissue-scale and systemic cues that are disrupted in aging. To fill in gaps between in vivo and traditional in vitro models, researchers have increasingly been turning to organotypic models, which provide increased physiological relevance with the accessibility and control of in vitro context. While powerful tools, the development of these models is a field of its own, and many aging researchers may be unaware of recent progress in organotypic models, or hesitant to include these models in their own work. In this review, we describe recent progress in tissue engineering applied to organotypic models, highlighting examples explicitly linked to aging and associated disease, as well as examples of models that are relevant to aging. We specifically highlight progress made in skin, gut, and skeletal muscle, and describe how recently demonstrated models have been used for aging studies or similar phenotypes. Throughout, this review emphasizes the accessibility of these models and aims to provide a resource for researchers seeking to leverage these powerful tools.
Collapse
Affiliation(s)
- Martina M. Sanchez
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - William Darch
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Joshua T. Morgan
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| |
Collapse
|
26
|
Huang Y, Gong Y, Liu Y, Lu J. Global trends and hot topics in electrical stimulation of skeletal muscle research over the past decade: A bibliometric analysis. Front Neurol 2022; 13:991099. [PMID: 36277916 PMCID: PMC9581161 DOI: 10.3389/fneur.2022.991099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/13/2022] [Indexed: 11/25/2022] Open
Abstract
Background Over the past decade, numerous advances have been made in the research on electrical stimulation of skeletal muscle. However, the developing status and future direction of this field remain unclear. This study aims to visualize the evolution and summarize global research hot topics and trends based on quantitative and qualitative evidence from bibliometrics. Methods Literature search was based on the Web of Science Core Collection (WoSCC) database from 2011 to 2021. CiteSpace and VOSviewer, typical bibliometric tools, were used to perform analysis and visualization. Results A total of 3,059 documents were identified. The number of literature is on the rise in general. Worldwide, researchers come primarily from North America and Europe, represented by the USA, France, Switzerland, and Canada. The Udice French Research Universities is the most published affiliation. Millet GY and Maffiuletti NA are the most prolific and the most co-cited authors, respectively. Plos One is the most popular journal, and the Journal of Applied Physiology is the top co-cited journal. The main keywords are muscle fatigue, neuromuscular electrical stimulation, spinal cord injury, tissue engineering, and atrophy. Moreover, this study systematically described the hotspots in this field. Conclusion As the first bibliometric analysis of electrical stimulation of skeletal muscle research over the past decade, this study can help scholars recognize hot topics and trends and provide a reference for further exploration in this field.
Collapse
|
27
|
Stalmach A, Boehm I, Fernandes M, Rutter A, Skipworth RJE, Husi H. Gene Ontology (GO)-Driven Inference of Candidate Proteomic Markers Associated with Muscle Atrophy Conditions. Molecules 2022; 27:5514. [PMID: 36080280 PMCID: PMC9457532 DOI: 10.3390/molecules27175514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle homeostasis is essential for the maintenance of a healthy and active lifestyle. Imbalance in muscle homeostasis has significant consequences such as atrophy, loss of muscle mass, and progressive loss of functions. Aging-related muscle wasting, sarcopenia, and atrophy as a consequence of disease, such as cachexia, reduce the quality of life, increase morbidity and result in an overall poor prognosis. Investigating the muscle proteome related to muscle atrophy diseases has a great potential for diagnostic medicine to identify (i) potential protein biomarkers, and (ii) biological processes and functions common or unique to muscle wasting, cachexia, sarcopenia, and aging alone. We conducted a meta-analysis using gene ontology (GO) analysis of 24 human proteomic studies using tissue samples (skeletal muscle and adipose biopsies) and/or biofluids (serum, plasma, urine). Whilst there were few similarities in protein directionality across studies, biological processes common to conditions were identified. Here we demonstrate that the GO analysis of published human proteomics data can identify processes not revealed by single studies. We recommend the integration of proteomics data from tissue samples and biofluids to yield a comprehensive overview of the human skeletal muscle proteome. This will facilitate the identification of biomarkers and potential pathways of muscle-wasting conditions for use in clinics.
Collapse
Affiliation(s)
- Angelique Stalmach
- Centre for Health Science, Division of Biomedical Sciences, Institute of Health Research and Innovation, School of Health, Social Care and Life Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
| | - Ines Boehm
- Edinburgh Cancer Research UK Tissue Group, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK
- Clinical Surgery, University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Marco Fernandes
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Alison Rutter
- Centre for Health Science, Division of Biomedical Sciences, Institute of Health Research and Innovation, School of Health, Social Care and Life Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
| | - Richard J. E. Skipworth
- Clinical Surgery, University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
| | - Holger Husi
- Centre for Health Science, Division of Biomedical Sciences, Institute of Health Research and Innovation, School of Health, Social Care and Life Sciences, University of the Highlands and Islands, Inverness IV2 3JH, UK
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| |
Collapse
|
28
|
Vann CG, Zhang X, Khodabukus A, Orenduff MC, Chen YH, Corcoran DL, Truskey GA, Bursac N, Kraus VB. Differential microRNA profiles of intramuscular and secreted extracellular vesicles in human tissue-engineered muscle. Front Physiol 2022; 13:937899. [PMID: 36091396 PMCID: PMC9452896 DOI: 10.3389/fphys.2022.937899] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Exercise affects the expression of microRNAs (miR/s) and muscle-derived extracellular vesicles (EVs). To evaluate sarcoplasmic and secreted miR expression in human skeletal muscle in response to exercise-mimetic contractile activity, we utilized a three-dimensional tissue-engineered model of human skeletal muscle ("myobundles"). Myobundles were subjected to three culture conditions: no electrical stimulation (CTL), chronic low frequency stimulation (CLFS), or intermittent high frequency stimulation (IHFS) for 7 days. RNA was isolated from myobundles and from extracellular vesicles (EVs) secreted by myobundles into culture media; miR abundance was analyzed by miRNA-sequencing. We used edgeR and a within-sample design to evaluate differential miR expression and Pearson correlation to evaluate correlations between myobundle and EV populations within treatments with statistical significance set at p < 0.05. Numerous miRs were differentially expressed between myobundles and EVs; 116 miRs were differentially expressed within CTL, 3 within CLFS, and 2 within IHFS. Additionally, 25 miRs were significantly correlated (18 in CTL, 5 in CLFS, 2 in IHFS) between myobundles and EVs. Electrical stimulation resulted in differential expression of 8 miRs in myobundles and only 1 miR in EVs. Several KEGG pathways, known to play a role in regulation of skeletal muscle, were enriched, with differentially overrepresented miRs between myobundle and EV populations identified using miEAA. Together, these results demonstrate that in vitro exercise-mimetic contractile activity of human engineered muscle affects both their expression of miRs and number of secreted EVs. These results also identify novel miRs of interest for future studies of the role of exercise in organ-organ interactions in vivo.
Collapse
Affiliation(s)
- Christopher G Vann
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Xin Zhang
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Alastair Khodabukus
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Melissa C. Orenduff
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Yu-Hsiu Chen
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - David L. Corcoran
- Department of Genetics, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - George A. Truskey
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Virginia B. Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Duke University, Durham, NC, United States
- Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
29
|
Vance DE, Lee Y, Batey DS, Li W, Chapman Lambert C, Nakkina SR, Anderson JN, Triebel K, Byun JY, Fazeli PL. Emerging directions of cognitive aging with HIV: practice and policy implications for social work. JOURNAL OF GERONTOLOGICAL SOCIAL WORK 2022; 65:476-494. [PMID: 34511048 DOI: 10.1080/01634372.2021.1978028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 06/13/2023]
Abstract
Cognitive impairments have been endemic to the HIV epidemic since its beginning and persist to this day. These impairments are attributed to HIV-induced neuroinflammation, the long-term effects of combination antiretroviral therapy, lifestyle factors (e.g., sedentary behavior, substance use), neuro-comorbidities (e.g., depression), age-associated comorbidities (e.g., heart disease, hypertension), and others causes. Normal aging and lifestyle also contribute to the development of cognitive impairment. Regardless of the etiology, such cognitive impairments interfere with HIV care (e.g., medication adherence) and everyday functioning (e.g., driving safely, financial management). With more than half of people with HIV (PWH) 50 years and older, and ~45% of all PWH meeting the criteria for HIV-Associated Neurocognitive Disorder (HAND), those aging PWH are more vulnerable for developing cognitive impairment. This article provides an update to a social work model to identify and monitor PWH for cognitive impairment. Within this update, the state of the science on protecting brain health and cognitive reserve within the context of neuroHIV is also presented. From this, implications for practice and policy to promote successful cognitive functioning in older PWH are provided.
Collapse
Affiliation(s)
- David E Vance
- School of Nursing, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yookyong Lee
- Department of Social Work, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David Scott Batey
- Department of Social Work, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei Li
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Sai Rashmi Nakkina
- College of Arts and Science, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joseph N Anderson
- School of Medicine, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kristen Triebel
- School of Medicine, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jun Y Byun
- School of Nursing, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Pariya L Fazeli
- School of Nursing, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
30
|
Wang J, Broer T, Chavez T, Zhou CJ, Tran S, Xiang Y, Khodabukus A, Diao Y, Bursac N. Myoblast deactivation within engineered human skeletal muscle creates a transcriptionally heterogeneous population of quiescent satellite-like cells. Biomaterials 2022; 284:121508. [PMID: 35421801 PMCID: PMC9289780 DOI: 10.1016/j.biomaterials.2022.121508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022]
Abstract
Satellite cells (SCs), the adult Pax7-expressing stem cells of skeletal muscle, are essential for muscle repair. However, in vitro investigations of SC function are challenging due to isolation-induced SC activation, loss of native quiescent state, and differentiation to myoblasts. In the present study, we optimized methods to deactivate in vitro expanded human myoblasts within a 3D culture environment of engineered human skeletal muscle tissues ("myobundles"). Immunostaining and gene expression analyses revealed that a fraction of myoblasts within myobundles adopted a quiescent phenotype (3D-SCs) characterized by increased Pax7 expression, cell cycle exit, and activation of Notch signaling. Similar to native SCs, 3D-SC quiescence is regulated by Notch and Wnt signaling while loss of quiescence and reactivation of 3D-SCs can be induced by growth factors including bFGF. Myobundle injury with a bee toxin, melittin, induces robust myofiber fragmentation, functional decline, and 3D-SC proliferation. By applying single cell RNA-sequencing (scRNA-seq), we discover the existence of two 3D-SC subpopulations (quiescent and activated), identify deactivation-associated gene signature using trajectory inference between 2D myoblasts and 3D-SCs, and characterize the transcriptomic changes within reactivated 3D-SCs in response to melittin-induced injury. These results demonstrate the ability of an in vitro engineered 3D human skeletal muscle environment to support the formation of a quiescent and heterogeneous SC population recapitulating several aspects of the native SC phenotype, and provide a platform for future studies of human muscle regeneration and disease-associated SC dysfunction.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Torie Broer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
31
|
Barthélémy F, Santoso JW, Rabichow L, Jin R, Little I, Nelson SF, McCain ML, Miceli MC. Modeling Patient-Specific Muscular Dystrophy Phenotypes and Therapeutic Responses in Reprogrammed Myotubes Engineered on Micromolded Gelatin Hydrogels. Front Cell Dev Biol 2022; 10:830415. [PMID: 35465312 PMCID: PMC9020228 DOI: 10.3389/fcell.2022.830415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
In vitro models of patient-derived muscle allow for more efficient development of genetic medicines for the muscular dystrophies, which often present mutation-specific pathologies. One popular strategy to generate patient-specific myotubes involves reprogramming dermal fibroblasts to a muscle lineage through MyoD induction. However, creating physiologically relevant, reproducible tissues exhibiting multinucleated, aligned myotubes with organized striations is dependent on the introduction of physicochemical cues that mimic the native muscle microenvironment. Here, we engineered patient-specific control and dystrophic muscle tissues in vitro by culturing and differentiating MyoD–directly reprogrammed fibroblasts isolated from one healthy control subject, three patients with Duchenne muscular dystrophy (DMD), and two Limb Girdle 2A/R1 (LGMD2A/R1) patients on micromolded gelatin hydrogels. Engineered DMD and LGMD2A/R1 tissues demonstrated varying levels of defects in α-actinin expression and organization relative to control, depending on the mutation. In genetically relevant DMD tissues amenable to mRNA reframing by targeting exon 44 or 45 exclusion, exposure to exon skipping antisense oligonucleotides modestly increased myotube coverage and alignment and rescued dystrophin protein expression. These findings highlight the value of engineered culture substrates in guiding the organization of reprogrammed patient fibroblasts into aligned muscle tissues, thereby extending their value as tools for exploration and dissection of the cellular and molecular basis of genetic muscle defects, rescue, and repair.
Collapse
Affiliation(s)
- Florian Barthélémy
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeffrey W. Santoso
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Laura Rabichow
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rongcheng Jin
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Isaiah Little
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
| | - Stanley F. Nelson
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, United States
- *Correspondence: M. Carrie Miceli, ; Megan L. McCain,
| | - M. Carrie Miceli
- Department of Microbiology Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: M. Carrie Miceli, ; Megan L. McCain,
| |
Collapse
|
32
|
Zschüntzsch J, Meyer S, Shahriyari M, Kummer K, Schmidt M, Kummer S, Tiburcy M. The Evolution of Complex Muscle Cell In Vitro Models to Study Pathomechanisms and Drug Development of Neuromuscular Disease. Cells 2022; 11:1233. [PMID: 35406795 PMCID: PMC8997482 DOI: 10.3390/cells11071233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Many neuromuscular disease entities possess a significant disease burden and therapeutic options remain limited. Innovative human preclinical models may help to uncover relevant disease mechanisms and enhance the translation of therapeutic findings to strengthen neuromuscular disease precision medicine. By concentrating on idiopathic inflammatory muscle disorders, we summarize the recent evolution of the novel in vitro models to study disease mechanisms and therapeutic strategies. A particular focus is laid on the integration and simulation of multicellular interactions of muscle tissue in disease phenotypes in vitro. Finally, the requirements of a neuromuscular disease drug development workflow are discussed with a particular emphasis on cell sources, co-culture systems (including organoids), functionality, and throughput.
Collapse
Affiliation(s)
- Jana Zschüntzsch
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Stefanie Meyer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Mina Shahriyari
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Karsten Kummer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Matthias Schmidt
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Susann Kummer
- Risk Group 4 Pathogens–Stability and Persistence, Biosafety Level-4 Laboratory, Center for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany;
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| |
Collapse
|
33
|
Samandari M, Quint J, Rodríguez-delaRosa A, Sinha I, Pourquié O, Tamayol A. Bioinks and Bioprinting Strategies for Skeletal Muscle Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105883. [PMID: 34773667 PMCID: PMC8957559 DOI: 10.1002/adma.202105883] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/28/2021] [Indexed: 05/16/2023]
Abstract
Skeletal muscles play important roles in critical body functions and their injury or disease can lead to limitation of mobility and loss of independence. Current treatments result in variable functional recovery, while reconstructive surgery, as the gold-standard approach, is limited due to donor shortage, donor-site morbidity, and limited functional recovery. Skeletal muscle tissue engineering (SMTE) has generated enthusiasm as an alternative solution for treatment of injured tissue and serves as a functional disease model. Recently, bioprinting has emerged as a promising tool for recapitulating the complex and highly organized architecture of skeletal muscles at clinically relevant sizes. Here, skeletal muscle physiology, muscle regeneration following injury, and current treatments following muscle loss are discussed, and then bioprinting strategies implemented for SMTE are critically reviewed. Subsequently, recent advancements that have led to improvement of bioprinting strategies to construct large muscle structures, boost myogenesis in vitro and in vivo, and enhance tissue integration are discussed. Bioinks for muscle bioprinting, as an essential part of any bioprinting strategy, are discussed, and their benefits, limitations, and areas to be improved are highlighted. Finally, the directions the field should expand to make bioprinting strategies more translational and overcome the clinical unmet needs are discussed.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - Olivier Pourquié
- Department of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
34
|
Jalal S, Dastidar S, Tedesco FS. Advanced models of human skeletal muscle differentiation, development and disease: Three-dimensional cultures, organoids and beyond. Curr Opin Cell Biol 2021; 73:92-104. [PMID: 34384976 PMCID: PMC8692266 DOI: 10.1016/j.ceb.2021.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/23/2021] [Indexed: 02/08/2023]
Abstract
Advanced in vitro models of human skeletal muscle tissue are increasingly needed to model complex developmental dynamics and disease mechanisms not recapitulated in animal models or in conventional monolayer cell cultures. There has been impressive progress towards creating such models by using tissue engineering approaches to recapitulate a range of physical and biochemical components of native human skeletal muscle tissue. In this review, we discuss recent studies focussed on developing complex in vitro models of human skeletal muscle beyond monolayer cell cultures, involving skeletal myogenic differentiation from human primary myoblasts or pluripotent stem cells, often in the presence of structural scaffolding support. We conclude with our outlook on the future of advanced skeletal muscle three-dimensional cultures (e.g. organoids and biofabrication) to produce physiologically and clinically relevant platforms for disease modelling and therapy development in musculoskeletal and neuromuscular disorders.
Collapse
Affiliation(s)
- Salma Jalal
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, United Kingdom
| | - Sumitava Dastidar
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, United Kingdom
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom; Department of Paediatric Neurology, Great Ormond Street Hospital for Children, WC1N 3JH London, United Kingdom.
| |
Collapse
|
35
|
Cho S, Jang J. Recent Trends in Biofabrication Technologies for Studying Skeletal Muscle Tissue-Related Diseases. Front Bioeng Biotechnol 2021; 9:782333. [PMID: 34778240 PMCID: PMC8578921 DOI: 10.3389/fbioe.2021.782333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
In native skeletal muscle, densely packed myofibers exist in close contact with surrounding motor neurons and blood vessels, which are embedded in the fibrous connective tissue. In comparison to conventional two-dimensional (2D) cultures, the three-dimensional (3D) engineered skeletal muscle models allow structural and mechanical resemblance with native skeletal muscle tissue by providing geometric confinement and physiological matrix stiffness to the cells. In addition, various external stimuli applied to these models enhance muscle maturation along with cell-cell and cell-extracellular matrix interaction. Therefore, 3D in vitro muscle models can adequately recapitulate the pathophysiologic events occurring in tissue-tissue interfaces inside the native skeletal muscle such as neuromuscular junction. Moreover, 3D muscle models can induce pathological phenotype of human muscle dystrophies such as Duchenne muscular dystrophy by incorporating patient-derived induced pluripotent stem cells and human primary cells. In this review, we discuss the current biofabrication technologies for modeling various skeletal muscle tissue-related diseases (i.e., muscle diseases) including muscular dystrophies and inflammatory muscle diseases. In particular, these approaches would enable the discovery of novel phenotypic markers and the mechanism study of human muscle diseases with genetic mutations.
Collapse
Affiliation(s)
- Seungyeun Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea
| |
Collapse
|
36
|
Chen Z, Anandakrishnan N, Xu Y, Zhao R. Compressive Buckling Fabrication of 3D Cell-Laden Microstructures. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101027. [PMID: 34263550 PMCID: PMC8425919 DOI: 10.1002/advs.202101027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/19/2021] [Indexed: 06/13/2023]
Abstract
Tissue architecture is a prerequisite for its biological functions. Recapitulating the three-dimensional (3D) tissue structure represents one of the biggest challenges in tissue engineering. Two-dimensional (2D) tissue fabrication methods are currently in the main stage for tissue engineering and disease modeling. However, due to their planar nature, the created models only represent very limited out-of-plane tissue structure. Here compressive buckling principle is harnessed to create 3D biomimetic cell-laden microstructures from microfabricated planar patterns. This method allows out-of-plane delivery of cells and extracellular matrix patterns with high spatial precision. As a proof of principle, a variety of polymeric 3D miniature structures including a box, an octopus, a pyramid, and continuous waves are fabricated. A mineralized bone tissue model with spatially distributed cell-laden lacunae structures is fabricated to demonstrate the fabrication power of the method. It is expected that this novel approach will help to significantly expand the utility of the established 2D fabrication techniques for 3D tissue fabrication. Given the widespread of 2D fabrication methods in biomedical research and the high demand for biomimetic 3D structures, this method is expected to bridge the gap between 2D and 3D tissue fabrication and open up new possibilities in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zhaowei Chen
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Nanditha Anandakrishnan
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Ying Xu
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Ruogang Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY, 14260, USA
| |
Collapse
|
37
|
Spotlight on Three Rs Progress. Altern Lab Anim 2021; 49:63-65. [PMID: 34225459 DOI: 10.1177/02611929211029118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Preclinical assessment on neuronal regeneration in the injury-related microenvironment of graphene-based scaffolds. NPJ Regen Med 2021; 6:31. [PMID: 34078912 PMCID: PMC8172906 DOI: 10.1038/s41536-021-00142-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/14/2021] [Indexed: 11/23/2022] Open
Abstract
As the application of graphene nanomaterials gets increasingly attractive in the field of tissue engineering and regenerative medicine, the long-term evaluation is necessary and urgent as to their biocompatibility and regenerative capacity in different tissue injuries, such as nerve, bone, and heart. However, it still remains controversial about the potential biological effects of graphene on neuronal activity, especially after severe nerve injuries. In this study, we establish a lengthy peripheral nerve defect rat model and investigate the potential toxicity of layered graphene-loaded polycaprolactone scaffold after implantation during 18 months in vivo. In addition, we further identify possible biologically regenerative effects of this scaffold on myelination, axonal outgrowth, and locomotor function recovery. It is confirmed that graphene-based nanomaterials exert negligible toxicity and repair large nerve defects by dual regulation of Schwann cells and astroglia in the central and peripheral nervous systems. The findings enlighten the future of graphene nanomaterial as a key type of biomaterials for clinical translation in neuronal regeneration.
Collapse
|
39
|
Clarke J. Exercise exerts anti-inflammatory effects on muscle via the JAK-STAT pathway. Nat Rev Rheumatol 2021; 17:127. [PMID: 33558742 DOI: 10.1038/s41584-021-00581-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|