1
|
Li W, Chen M, Zhang L. Muscle Stem Cell Microenvironment and Functions in Muscle Regeneration. Biomolecules 2025; 15:765. [PMID: 40563407 PMCID: PMC12190381 DOI: 10.3390/biom15060765] [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: 04/02/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 06/28/2025] Open
Abstract
Muscle stem cells (MuSCs) are the key to muscle regeneration. The activation and maintenance of MuSCs require the precise regulation of their microenvironments. Myofibers and other cells including endothelial cells, fibroblasts, and immune cell populations constitute the cell components of the MuSC niche. The communication between these cell populations and MuSCs play an essential role in muscle repair. Furthermore, the physical and chemical stimulations around MuSCs also affect the cell behaviors of MuSCs. Extracellular matrix (ECM) and the factors stored in it generate a repair-promoting niche for efficient muscle regeneration. Understanding the mechanism of muscle stem cell regulation is the basis of clinically optimizing muscle repair. In this review, we discuss recent findings about the microenvironments of MuSCs and their functions in muscle regeneration, which would shed light on new targets and strategies for muscle injury treatment.
Collapse
Affiliation(s)
- Wenjing Li
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| | - Minyou Chen
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China;
| | - Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China;
| |
Collapse
|
2
|
Marjan T, Lafuente-Gómez N, Rampal A, Mooney DJ, Peyton SR, Qazi TH. Cell-Instructive Biomaterials with Native-Like Biochemical Complexity. Annu Rev Biomed Eng 2025; 27:185-209. [PMID: 39874600 PMCID: PMC12045723 DOI: 10.1146/annurev-bioeng-120823-020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Biochemical signals in native tissue microenvironments instruct cell behavior during many biological processes ranging from developmental morphogenesis and tissue regeneration to tumor metastasis and disease progression. The detection and characterization of these signals using spatial and highly resolved quantitative methods have revealed their existence as matricellular proteins in the matrisome, some of which are bound to the extracellular matrix while others are freely diffusing. Including these biochemical signals in engineered biomaterials can impart enhanced functionality and native-like complexity, ultimately benefiting efforts to understand, model, and treat various diseases. In this review, we discuss advances in characterizing, mimicking, and harnessing biochemical signals in developing advanced engineered biomaterials. An overview of the diverse forms in which these biochemical signals exist and their effects on intracellular signal transduction is also provided. Finally, we highlight the application of biochemically complex biomaterials in the three broadly defined areas of tissue regeneration, immunoengineering, and organoid morphogenesis.
Collapse
Affiliation(s)
- Tuba Marjan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Nuria Lafuente-Gómez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Akaansha Rampal
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA;
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA;
| | - Taimoor H Qazi
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA;
| |
Collapse
|
3
|
Liang T, Zhang H, Hu Y, Solanki M, Zhang C, Sasaki T, Smith CE, Hu JCC, Simmer JP. Localizations of Laminin Chains Suggest Their Multifaceted Functions in Mouse Tooth Development. Int J Mol Sci 2025; 26:4134. [PMID: 40362374 PMCID: PMC12071823 DOI: 10.3390/ijms26094134] [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: 02/28/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
The human laminin family is composed of five α, four β, and three γ chains. Laminins are heterotrimers of α, β, and γ chains. Laminins play critical roles during organogenesis, mostly as basement membrane components. The expression of all and the localization of most laminin chains were characterized in mouse developing teeth. Primary laminin isoforms in basement membranes along the inner enamel epithelium before the secretory stage and outside of the outer enamel epithelium were laminins 111 (α1β1γ1) and 511. The mouse laminin α3 chain has two variants, α3A and α3B. Although a basement membrane structure is absent, laminin 3A32 was localized along the secretory surface of the secretory stage ameloblast Tomes' processes. Laminin 3A32 was localized along the atypical basement membrane of maturation stage ameloblasts and the specialized basement membrane of junctional epithelium facing the enamel surface. The endothelial basement membrane in the dental papilla and outside of the enamel organ contained laminins 411 and 511. Laminin 332 was detected in the extracellular matrix but not the basement membrane of the apical loop. Laminin 111 was localized in the extracellular matrix of the apical dental papilla without forming a visible basement membrane. These findings suggest the multifaceted functions of laminins in tooth development and set the foundation for functional investigations.
Collapse
Affiliation(s)
- Tian Liang
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (Y.H.); (M.S.)
| | - Hong Zhang
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (H.Z.); (C.Z.); (C.E.S.); (J.C.-C.H.); (J.P.S.)
| | - Yuanyuan Hu
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (Y.H.); (M.S.)
| | - Mansi Solanki
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (Y.H.); (M.S.)
| | - Chuhua Zhang
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (H.Z.); (C.Z.); (C.E.S.); (J.C.-C.H.); (J.P.S.)
| | - Takako Sasaki
- Department of Matrix Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-Machi, Yufu City 879-5593, Oita, Japan;
| | - Charles E. Smith
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (H.Z.); (C.Z.); (C.E.S.); (J.C.-C.H.); (J.P.S.)
- Department of Anatomy & Cell Biology, Faculty of Medicine & Health Sciences, McGill University, 3640 University St., Montreal, QC H3A 0C7, Canada
| | - Jan C.-C. Hu
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (H.Z.); (C.Z.); (C.E.S.); (J.C.-C.H.); (J.P.S.)
| | - James P. Simmer
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, 1011 North University, Ann Arbor, MI 48109, USA; (H.Z.); (C.Z.); (C.E.S.); (J.C.-C.H.); (J.P.S.)
| |
Collapse
|
4
|
Park G, Grey JA, Mourkioti F, Han WM. 3D Mechanical Confinement Directs Muscle Stem Cell Fate and Function. Adv Biol (Weinh) 2025; 9:e2400717. [PMID: 40040295 PMCID: PMC12001014 DOI: 10.1002/adbi.202400717] [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: 11/17/2024] [Revised: 02/07/2025] [Indexed: 03/06/2025]
Abstract
Muscle stem cells (MuSCs) play a crucial role in skeletal muscle regeneration, residing in a niche that undergoes dimensional and mechanical changes throughout the regeneration process. This study investigates how 3D confinement and stiffness encountered by MuSCs during the later stages of regeneration regulate their function, including stemness, activation, proliferation, and differentiation. An asymmetric 3D hydrogel bilayer platform is engineered with tunable physical constraints to mimic the regenerating MuSC niche. These results demonstrate that increased 3D confinement maintains Pax7 expression, reduces MuSC activation and proliferation, inhibits differentiation, and is associated with smaller nuclear size and decreased H4K16ac levels, suggesting that mechanical confinement modulates both nuclear architecture and epigenetic regulation. MuSCs in unconfined 2D environments exhibit larger nuclei and higher H4K16ac expression compared to those in more confined 3D conditions, leading to progressive activation, expansion, and myogenic commitment. This study highlights the importance of 3D mechanical cues in MuSC fate regulation, with 3D confinement acting as a mechanical brake on myogenic commitment, offering novel insights into the mechano-epigenetic mechanisms that govern MuSC behavior during muscle regeneration.
Collapse
Affiliation(s)
- GaYoung Park
- Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Josh A. Grey
- Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of CellDevelopmentand Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Institute for Regenerative MedicineIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Foteini Mourkioti
- Department of Orthopaedic SurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Cell and Developmental BiologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Institute for Regenerative MedicineMusculoskeletal ProgramPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Woojin M. Han
- Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of CellDevelopmentand Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Institute for Regenerative MedicineIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| |
Collapse
|
5
|
Fancello I, Willett S, Castiglioni C, Amer S, Santoleri S, Bragg L, Galli F, Cossu G. TNAP expressing adventitial pericytes contribute to myogenesis during foetal development. Vascul Pharmacol 2025; 159:107489. [PMID: 40097085 DOI: 10.1016/j.vph.2025.107489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE During growth and differentiation of skeletal muscle, cell types other than canonical myoblasts can be recruited to a myogenic fate. Among these, TNAP+ pericytes can differentiate into skeletal or smooth muscle cells during postnatal growth and contribute to muscle regeneration. However, their role in muscle development has not been investigated. This study aims to characterise pericyte fate choices during embryonic and foetal myogenesis, occurring in the second half of gestation. APPROACH AND RESULTS Using Cre-loxP lineage tracing with multiple reporters including the multifluorescent Confetti, we labelled TNAP+ precursors in vivo and assessed the smooth or skeletal muscle differentiation in their lineage at a perinatal stage. We found that TNAP+ cells contribute in vivo to skeletal and smooth muscle cells, as well as other pericytes, also during pre-natal muscle development. The resulting clones showed that such fate choices are likely to depend on distinct unipotent progenitors rather than multipotent progenitors. In addition, we isolated and differentiated in vitro foetal cells derived from TNAP+ precursors, which showed that they are not spontaneously myogenic unless co-cultured with other skeletal muscle cells. CONCLUSIONS This work extends our understanding of the differentiative potency of these non- canonical skeletal muscle progenitors during prenatal life, with a view to a future application of this knowledge to optimise cell therapies for muscle wasting disorders.
Collapse
Affiliation(s)
- I Fancello
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Willett
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - C Castiglioni
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Amer
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Santoleri
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - L Bragg
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - F Galli
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - G Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK; Institute of Experimental Neurology, Division of Neurosciences, Ospedale San Raffaele, Milan, Italy; Experimental and Clinical Research Center, Charité Medical Faculty, Max Delbrück Center Berlin, Germany.
| |
Collapse
|
6
|
Then AA, Goenawan H, Lesmana R, Christoper A, Sylviana N, Gunadi JW. Exploring the potential regulation of DUOX in thyroid hormone‑autophagy signaling via IGF‑1 in the skeletal muscle (Review). Biomed Rep 2025; 22:39. [PMID: 39781041 PMCID: PMC11704872 DOI: 10.3892/br.2024.1917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/16/2024] [Indexed: 01/11/2025] Open
Abstract
Dual oxidases (DUOX) are enzymes that have the main function in producing reactive oxygen species (ROS) in various tissues. DUOX also play an important role in the synthesis of H2O2, which is essential for the production of thyroid hormone. Thyroid hormones can influence the process of muscle development through direct stimulation of ROS, 5' AMP-activated protein kinase (AMPK) and mTOR and indirect effect autophagy and the insulin-like growth factor 1 (IGF-1) pathway. IGF-1 signaling controls autophagy in two ways: Inhibiting autophagy through activation of the PI3K/AKT/mTOR/MAPK pathway and promoting mitophagy through the nuclear factor erythroid 2-related factor 2-binding receptor Bcl2/adenovirus E1B 19 kDa protein-interacting protein 3. Thyroid hormone deficiency caused by the absence of DUOX should be considered because it might have a significant effect on the growth of skeletal muscle. The effect of DUOX regulation on thyroid hormone autophagy via IGF-1 in skeletal muscle has not been well investigated. The present review discussed the regulatory interactions between DUOX, thyroid hormone, IGF-1 and autophagy, which can influence skeletal muscle development.
Collapse
Affiliation(s)
- Andreas Adiwinata Then
- Master's Program in Basic Biomedical Sciences, Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java 40161, Indonesia
| | - Hanna Goenawan
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor-Sumedang, West Java 45363, Indonesia
- Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Jatinangor-Sumedang, West Java 45363, Indonesia
| | - Ronny Lesmana
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor-Sumedang, West Java 45363, Indonesia
- Biological Activity Division, Central Laboratory, Universitas Padjadjaran, Jatinangor-Sumedang, West Java 45363, Indonesia
| | - Andreas Christoper
- Doctoral Program in Medical Science, PMDSU Program Batch VI, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java 40161, Indonesia
| | - Nova Sylviana
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor-Sumedang, West Java 45363, Indonesia
| | - Julia Windi Gunadi
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java 40164, Indonesia
| |
Collapse
|
7
|
Barajaa MA, Ghosh D, Laurencin CT. Decellularized Extracellular Matrix-Derived Hydrogels: a Powerful Class of Biomaterials for Skeletal Muscle Regenerative Engineering Applications. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2025; 11:39-63. [PMID: 40201194 PMCID: PMC11978403 DOI: 10.1007/s40883-023-00328-8] [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: 08/23/2023] [Revised: 10/18/2023] [Accepted: 11/28/2023] [Indexed: 04/10/2025]
Abstract
Purpose The extracellular matrix (ECM) is a complicated milieu consisting of structural and functional molecules secreted by the resident cells that provides an optimal microenvironmental niche for enhanced cell adhesion, growth, differentiation, and tissue formation and maturation. For decades, ECM bio-scaffolds prepared from decellularized tissues have been used to promote skeletal muscle regeneration; however, it was recently discovered that these decellularized ECM (dECM) materials can be further processed into hydrogels, thus expanding the potential applications of dECM materials in skeletal muscle regenerative engineerisng (SMRE). This review article highlights the recent advances in dECM-derived hydrogels toward skeletal muscle regeneration and repair. Method We screened articles in PubMed and bibliographic search using a combination of keywords. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Here, we discuss the skeletal muscle ECM's structure, function, and biochemical composition with emphasis on the role of the ECM during skeletal muscle embryogenesis, growth, development, and repair. Furthermore, we review various hydrogels used to promote skeletal muscle regeneration. We also review the current applications of dECM-derived hydrogels toward SMRE. Finally, we discuss the clinical translation potential of dECM-derived hydrogels for skeletal muscle regeneration and repair and their potential clinical considerations in the future. Conclusion Although much progress has been made in the field of dECM-derived hydrogels toward SMRE, it is still in its nascent stage. We believe improving and standardizing the methods of decellularization, lowering the immunogenicity of dECMs, and carrying out in vivo investigations in large animal models would advance their future clinical applications. Lay Summary Researchers have discovered an effective way to turn tissue materials into jelly-like substances known as extracellular matrix (ECM)-derived hydrogels. These ECM-derived hydrogels can help muscles heal better after serious injuries. They can be injected into gaps or used to guide muscle growth in the lab or body. This review article explains how these ECM-derived hydrogels are made and how they can be used to improve muscle healing. It also discusses their possible use in clinics and what needs to be considered before using them for medical treatments.
Collapse
Affiliation(s)
- Mohammed A. Barajaa
- Department of Biomedical Engineering, College of Engineering, Imam Abdulrahman Bin Faisal University, 34212 Dammam, Saudi Arabia
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT 06030, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
8
|
Yamashita AMS, Garay BI, Kim H, Bosnakovski D, Abrahante JE, Azzag K, Abreu P, Ahlquist A, Perlingeiro RCR. Effect of Notch1 signaling on muscle engraftment and maturation from pluripotent stem cells. Stem Cell Reports 2025; 20:102396. [PMID: 39889709 PMCID: PMC11864150 DOI: 10.1016/j.stemcr.2024.102396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Pax3-induced pluripotent stem cell-derived myogenic progenitors display an embryonic molecular signature but become postnatal upon transplantation. Because this correlates with upregulation of Notch signaling, here we probed whether NOTCH1 is required for in vivo maturation by performing gain- and loss-of-function studies in inducible Pax3 (iPax3) myogenic progenitors. Transplantation studies revealed that Notch1 signaling did not change the number of donor-derived fibers; however, the NOTCH1 overexpression cohorts showed enhanced satellite cell engraftment and more mature fibers, as indicated by fewer fibers expressing the embryonic myosin heavy-chain isoform and more type IIX fibers. While donor-derived Pax7+ cells were detected in all transplants, in the absence of Notch1, secondary grafts exhibited a high fraction of these cells in the interstitial space, indicating that NOTCH1 is required for proper satellite cell homing. Transcriptional profiling of NOTCH1-modified donor-derived satellite cells suggests that this may be due to changes in the extracellular matrix organization, cell cycle, and metabolism.
Collapse
Affiliation(s)
- Aline M S Yamashita
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Bayardo I Garay
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Hyunkee Kim
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Darko Bosnakovski
- Lillehei Heart Institute, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Juan E Abrahante
- University of Minnesota Informatics Institute, Minneapolis, MN, USA
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Phablo Abreu
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Aaron Ahlquist
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
9
|
Chang TL, Borelli AN, Cutler AA, Olwin BB, Anseth KS. Myofibers cultured in viscoelastic hydrogels reveal the effects of integrin-binding and mechanosensing on muscle satellite cells. Acta Biomater 2025; 192:48-60. [PMID: 39615561 PMCID: PMC11949280 DOI: 10.1016/j.actbio.2024.11.044] [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: 07/23/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
Quiescent skeletal muscle satellite cells (SCs) located on myofibers activate in response to muscle injury to regenerate muscle; however, identifying the role of specific matrix signals on SC behavior in vivo is difficult. Therefore, we developed a viscoelastic hydrogel with tunable properties to encapsulate myofibers while maintaining stem cell niche polarity and SC-myofiber interactions to investigate how matrix signals, including viscoelasticity and the integrin-binding ligand arginyl-glycyl-aspartic acid (RGD), influence SC behavior during muscle regeneration. Viscoelastic hydrogels support myofiber culture while preserving SC stemness for up to 72 hours post-encapsulation, minimizing myofiber hypercontraction and SC hyperproliferation compared to Matrigel. Pax7 is continuously expressed in SCs on myofibers embedded in hydrogels with higher stress relaxation while SCs differentiate when embedded in elastic hydrogels. Increasing RGD concentrations activates SCs and translocates YAP/TAZ to the nucleus as revealed by photo-expansion microscopy. Deleting YAP/TAZ abrogates RGD-mediated activation of SCs, and thus, YAP/TAZ mediates RGD ligand-induced SC activation and subsequent proliferation. STATEMENT OF SIGNIFICANCE: Satellite cells (SCs) are responsible for muscle maintenance and regeneration, but how the extracellular matrix regulates SC function is less understood and would benefit from new biomaterial models that can recapitulate the complexity of SC niche in vitro. Upon isolation of myofibers, SCs exit quiescence, becoming activated. To circumvent this issue, we developed a viscoelastic hydrogel for encapsulating myofibers, which maintains SC quiescence and limits differentiation, allowing the study of RGD effects. We showed that increasing RGD concentration promotes activation and suppresses differentiation. Finally, to allow high resolution imaging for resolving the subcellular localization of YAP/TAZ transcriptional co-activators, we applied photo-expansion microscopy and gel-to-gel transfer techniques to quantify YAP/TAZ nuclear-cytoplasmic ratio, revealing that RGD-mediated activation relies on YAP/TAZ nuclear translocation.
Collapse
Affiliation(s)
- Tze-Ling Chang
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder CO, 80303, USA; The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Alexandra N Borelli
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder CO, 80303, USA; The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Alicia A Cutler
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Bradley B Olwin
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA; Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder CO, 80303, USA; The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
10
|
Mitra M, Batista SL, Coller HA. Transcription factor networks in cellular quiescence. Nat Cell Biol 2025; 27:14-27. [PMID: 39789221 DOI: 10.1038/s41556-024-01582-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025]
Abstract
Many of the cells in mammalian tissues are in a reversible quiescent state; they are not dividing, but retain the ability to proliferate in response to extracellular signals. Quiescence relies on the activities of transcription factors (TFs) that orchestrate the repression of genes that promote proliferation and establish a quiescence-specific gene expression program. Here we discuss how the coordinated activities of TFs in different quiescent stem cells and differentiated cells maintain reversible cell cycle arrest and establish cell-protective signalling pathways. We further cover the emerging mechanisms governing the dysregulation of quiescence TF networks with age. We explore how recent developments in single-cell technologies have enhanced our understanding of quiescence heterogeneity and gene regulatory networks. We further discuss how TFs and their activities are themselves regulated at the RNA, protein and chromatin levels. Finally, we summarize the challenges associated with defining TF networks in quiescent cells.
Collapse
Affiliation(s)
- Mithun Mitra
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Sandra L Batista
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Yin K, Zhang C, Deng Z, Wei X, Xiang T, Yang C, Chen C, Chen Y, Luo F. FAPs orchestrate homeostasis of muscle physiology and pathophysiology. FASEB J 2024; 38:e70234. [PMID: 39676717 PMCID: PMC11647758 DOI: 10.1096/fj.202400381r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024]
Abstract
As a common clinical manifestation, muscle weakness is prevalent in people with mobility disorders. Further studies of muscle weakness have found that patients with muscle weakness present with persistent muscle inflammation, loss of muscle fibers, fat infiltration, and interstitial fibrosis. Therefore, we propose the concept of muscle microenvironment homeostasis, which explains the abnormal pathological changes in muscles through the imbalance of muscle microenvironment homeostasis. And we identified an interstitial progenitor cell FAP during the transition from normal muscle microenvironment homeostasis to muscle microenvironment imbalance caused by muscle damage diseases. As a kind of pluripotent stem cell, FAPs do not participate in myogenic differentiation, but can differentiate into fibroblasts, adipocytes, osteoblasts, and chondrocytes. As a kind of mesenchymal progenitor cell, it is involved in the generation of extracellular matrix, regulate muscle regeneration, and maintain neuromuscular junction. However, the muscle microenvironment is disrupted by the causative factors, and the abnormal activities of FAPs eventually contribute to the complex pathological changes in muscles. Targeting the mechanisms of these muscle pathological changes, we have identified appropriate signaling targets for FAPs to improve and even treat muscle damage diseases. In this review, we propose the construction of muscle microenvironmental homeostasis and find the key cells that cause pathological changes in muscle after homeostasis is broken. By studying the mechanism of abnormal differentiation and apoptosis of FAPs, we found a strategy to inhibit the abnormal pathological changes in muscle damage diseases and improve muscle regeneration.
Collapse
Affiliation(s)
- Kai Yin
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Chengmin Zhang
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Zihan Deng
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Xiaoyu Wei
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Tingwen Xiang
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Chuan Yang
- Department of Biomedical Materials ScienceThird Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Can Chen
- Department for Combat Casualty Care TrainingTraining Base for Army Health Care, Army Medical University (Third Military Medical University)ChongqingPeople's Republic of China
| | - Yueqi Chen
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| | - Fei Luo
- Department of OrthopedicsSouthwest Hospital, Third Military Medical University (Army Medical University)ChongqingPeople's Republic of China
| |
Collapse
|
12
|
Sommers O, Tomsine RA, Khacho M. Mitochondrial Dynamics Drive Muscle Stem Cell Progression from Quiescence to Myogenic Differentiation. Cells 2024; 13:1773. [PMID: 39513880 PMCID: PMC11545319 DOI: 10.3390/cells13211773] [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: 09/29/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
From quiescence to activation and myogenic differentiation, muscle stem cells (MuSCs) experience drastic alterations in their signaling activity and metabolism. Through balanced cycles of fission and fusion, mitochondria alter their morphology and metabolism, allowing them to affect their decisive role in modulating MuSC activity and fate decisions. This tightly regulated process contributes to MuSC regulation by mediating changes in redox signaling pathways, cell cycle progression, and cell fate decisions. In this review, we discuss the role of mitochondrial dynamics as an integral modulator of MuSC activity, fate, and maintenance. Understanding the influence of mitochondrial dynamics in MuSCs in health and disease will further the development of therapeutics that support MuSC integrity and thus may aid in restoring the regenerative capacity of skeletal muscle.
Collapse
Affiliation(s)
- Olivia Sommers
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rholls A. Tomsine
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Center for Neuromuscular Disease (CNMD), University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
13
|
Mei J, Jiang XY, Tian HX, Rong DC, Song JN, Wang L, Chen YS, Wong RCB, Guo CX, Wang LS, Wang LY, Wang PY, Yin JY. Anoikis in cell fate, physiopathology, and therapeutic interventions. MedComm (Beijing) 2024; 5:e718. [PMID: 39286778 PMCID: PMC11401975 DOI: 10.1002/mco2.718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
The extracellular matrix (ECM) governs a wide spectrum of cellular fate processes, with a particular emphasis on anoikis, an integrin-dependent form of cell death. Currently, anoikis is defined as an intrinsic apoptosis. In contrast to traditional apoptosis and necroptosis, integrin correlates ECM signaling with intracellular signaling cascades, describing the full process of anoikis. However, anoikis is frequently overlooked in physiological and pathological processes as well as traditional in vitro research models. In this review, we summarized the role of anoikis in physiological and pathological processes, spanning embryonic development, organ development, tissue repair, inflammatory responses, cardiovascular diseases, tumor metastasis, and so on. Similarly, in the realm of stem cell research focused on the functional evolution of cells, anoikis offers a potential solution to various challenges, including in vitro cell culture models, stem cell therapy, cell transplantation, and engineering applications, which are largely based on the regulation of cell fate by anoikis. More importantly, the regulatory mechanisms of anoikis based on molecular processes and ECM signaling will provide new strategies for therapeutic interventions (drug therapy and cell-based therapy) in disease. In summary, this review provides a systematic elaboration of anoikis, thus shedding light on its future research.
Collapse
Affiliation(s)
- Jie Mei
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Xue-Yao Jiang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Hui-Xiang Tian
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Ding-Chao Rong
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
| | - Jia-Nan Song
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- School of Life Sciences Westlake University Hangzhou Zhejiang China
| | - Luozixian Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Yuan-Shen Chen
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Raymond C B Wong
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Cheng-Xian Guo
- Center of Clinical Pharmacology the Third Xiangya Hospital Central South University Changsha Hunan China
| | - Lian-Sheng Wang
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Lei-Yun Wang
- Department of Pharmacy Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei Province China
| | - Peng-Yuan Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
14
|
Madl CM, Wang YX, Holbrook CA, Su S, Shi X, Byfield FJ, Wicki G, Flaig IA, Blau HM. Hydrogel biomaterials that stiffen and soften on demand reveal that skeletal muscle stem cells harbor a mechanical memory. Proc Natl Acad Sci U S A 2024; 121:e2406787121. [PMID: 39163337 PMCID: PMC11363279 DOI: 10.1073/pnas.2406787121] [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: 04/04/2024] [Accepted: 07/21/2024] [Indexed: 08/22/2024] Open
Abstract
Muscle stem cells (MuSCs) are specialized cells that reside in adult skeletal muscle poised to repair muscle tissue. The ability of MuSCs to regenerate damaged tissues declines markedly with aging and in diseases such as Duchenne muscular dystrophy, but the underlying causes of MuSC dysfunction remain poorly understood. Both aging and disease result in dramatic increases in the stiffness of the muscle tissue microenvironment from fibrosis. MuSCs are known to lose their regenerative potential if cultured on stiff plastic substrates. We sought to determine whether MuSCs harbor a memory of their past microenvironment and if it can be overcome. We tested MuSCs in situ using dynamic hydrogel biomaterials that soften or stiffen on demand in response to light and found that freshly isolated MuSCs develop a persistent memory of substrate stiffness characterized by loss of proliferative progenitors within the first three days of culture on stiff substrates. MuSCs cultured on soft hydrogels had altered cytoskeletal organization and activity of Rho and Rac guanosine triphosphate hydrolase (GTPase) and Yes-associated protein mechanotransduction pathways compared to those on stiff hydrogels. Pharmacologic inhibition identified RhoA activation as responsible for the mechanical memory phenotype, and single-cell RNA sequencing revealed a molecular signature of the mechanical memory. These studies highlight that microenvironmental stiffness regulates MuSC fate and leads to MuSC dysfunction that is not readily reversed by changing stiffness. Our results suggest that stiffness can be circumvented by targeting downstream signaling pathways to overcome stem cell dysfunction in aged and disease states with aberrant fibrotic tissue mechanics.
Collapse
Affiliation(s)
- Christopher M. Madl
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA19104
| | - Yu Xin Wang
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
| | - Colin A. Holbrook
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
| | - Shiqi Su
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
| | - Xuechen Shi
- Department of Physiology, Perelman School of Medicine and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA19104
| | - Fitzroy J. Byfield
- Department of Physiology, Perelman School of Medicine and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA19104
| | - Gwendoline Wicki
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
- Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, LausanneCH-1015, Switzerland
| | - Iris A. Flaig
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Federale de Lausanne, LausanneCH-1015, Switzerland
| | - Helen M. Blau
- Department of Microbiology and Immunology, Baxter Laboratory for Stem Cell Biology, Stanford University, Stanford, CA94305
| |
Collapse
|
15
|
Khan HA, Van Hateren N, Borycki AG. Light-Sheet Microscopy Enables Three-Dimensional Fluorescence Imaging and Live Imaging of Satellite Cells on Skeletal Muscle Fibers. Methods Mol Biol 2024. [PMID: 38997538 DOI: 10.1007/7651_2024_552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
The ex vivo myofiber culture system has proven to be a useful methodology to explore the biology and behavior of satellite cells within their niche environment. However, a limitation of this system is that myofibers and their associated satellite cells are commonly examined using conventional fluorescence microscopy, which renders a three-dimensional system into two-dimensional imaging, leading to the loss of precious information or misleading interpretation of observations. Here, we report on the use of light-sheet fluorescence microscopy to generate three-dimensional and live imaging of satellite cells on myofibers. Light-sheet microscopy offers high imaging speed and good spatial resolution with minimal photo-bleaching, allowing live imaging and three-dimensional acquisition of skeletal muscle fiber specimen. The potentials of this technology are wide, ranging from the visualization of satellite cell behavior such as cell division and cell migration to imaging the sub-cellular localization of proteins or organelles.
Collapse
Affiliation(s)
- Hira Asif Khan
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Nick Van Hateren
- The Wolfson Light Microscopy Facility, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Anne-Gaëlle Borycki
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK.
| |
Collapse
|
16
|
Millward DJ. Post-natal muscle growth and protein turnover: a narrative review of current understanding. Nutr Res Rev 2024; 37:141-168. [PMID: 37395180 DOI: 10.1017/s0954422423000124] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
A model explaining the dietary-protein-driven post-natal skeletal muscle growth and protein turnover in the rat is updated, and the mechanisms involved are described, in this narrative review. Dietary protein controls both bone length and muscle growth, which are interrelated through mechanotransduction mechanisms with muscle growth induced both from stretching subsequent to bone length growth and from internal work against gravity. This induces satellite cell activation, myogenesis and remodelling of the extracellular matrix, establishing a growth capacity for myofibre length and cross-sectional area. Protein deposition within this capacity is enabled by adequate dietary protein and other key nutrients. After briefly reviewing the experimental animal origins of the growth model, key concepts and processes important for growth are reviewed. These include the growth in number and size of the myonuclear domain, satellite cell activity during post-natal development and the autocrine/paracrine action of IGF-1. Regulatory and signalling pathways reviewed include developmental mechanotransduction, signalling through the insulin/IGF-1-PI3K-Akt and the Ras-MAPK pathways in the myofibre and during mechanotransduction of satellite cells. Likely pathways activated by maximal-intensity muscle contractions are highlighted and the regulation of the capacity for protein synthesis in terms of ribosome assembly and the translational regulation of 5-TOPmRNA classes by mTORC1 and LARP1 are discussed. Evidence for and potential mechanisms by which volume limitation of muscle growth can occur which would limit protein deposition within the myofibre are reviewed. An understanding of how muscle growth is achieved allows better nutritional management of its growth in health and disease.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, School of Biosciences & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
17
|
Duran P, Yang BA, Plaster E, Eiken M, Loebel C, Aguilar CA. Tracking of Nascent Matrix Deposition during Muscle Stem Cell Activation across Lifespan Using Engineered Hydrogels. Adv Biol (Weinh) 2024; 8:e2400091. [PMID: 38616175 DOI: 10.1002/adbi.202400091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/22/2024] [Indexed: 04/16/2024]
Abstract
Adult stem cells occupy a niche that contributes to their function, but how stem cells rebuild their microenvironment after injury remains an open-ended question. Herein, biomaterial-based systems and metabolic labeling are utilized to evaluate how skeletal muscle stem cells deposit extracellular matrix. Muscle stem cells and committed myoblasts are observed to generate less nascent matrix than muscle resident fibro-adipogenic progenitors. When cultured on substrates that matched the stiffness of physiological uninjured and injured muscles, muscle stem cells increased nascent matrix deposition with activation kinetics. Reducing the ability to deposit nascent matrix by an inhibitor of vesicle trafficking (Exo-1) attenuated muscle stem cell function and mimicked impairments observed from muscle stem cells isolated from old muscles. Old muscle stem cells are observed to deposit less nascent matrix than young muscle stem cells, which is rescued with therapeutic supplementation of insulin-like growth factors. These results highlight the role of nascent matrix production with muscle stem cell activation.
Collapse
Affiliation(s)
- Pamela Duran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Benjamin A Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eleanor Plaster
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Madeline Eiken
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Carlos A Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
18
|
Louie JD, Barrios-Camacho CM, Bromberg BH, Hintschich CA, Schwob JE. Spatiotemporal dynamics exhibited by horizontal basal cells reveal a pro-neurogenic pathway during injury-induced olfactory epithelium regeneration. iScience 2024; 27:109600. [PMID: 38650985 PMCID: PMC11033173 DOI: 10.1016/j.isci.2024.109600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 12/21/2023] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Horizontal basal cells (HBCs) mediate olfactory epithelium (OE) regeneration following severe tissue injury. The dynamism of the post-injury environment is well illustrated by in silico modeling of RNA sequencing data that demonstrate an evolving HBC transcriptome. Unfortunately, spatiotemporally dynamic processes occurring within HBCs in situ remain poorly understood. Here, we show that HBCs at 24 h post-OE injury spatially redistribute a constellation of proteins, which, in turn, helped to nominate Rac1 as a regulator of HBC differentiation during OE regeneration. Using our primary culture model to activate HBCs pharmacologically, we demonstrate that concurrent Rac1 inhibition attenuates HBC differentiation potential. This in vitro functional impairment manifested in vivo as decreased HBC differentiation into olfactory sensory neurons following HBC-specific Rac1 conditional knockout. Taken together, our data potentiate the design of hyposmia-alleviating therapies and highlight aspects of in situ HBC spatiotemporal dynamics that deserve further investigation.
Collapse
Affiliation(s)
- Jonathan D. Louie
- Medical Scientist Training Program, Tufts University School of Medicine, Boston, MA 02111, USA
- Neuroscience Graduate Program, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
- Department of Developmental, Molecular & Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Camila M. Barrios-Camacho
- Neuroscience Graduate Program, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
- Department of Developmental, Molecular & Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Benjamin H. Bromberg
- Department of Developmental, Molecular & Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Constantin A. Hintschich
- Department of Developmental, Molecular & Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
- Department of Otorhinolaryngology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - James E. Schwob
- Department of Developmental, Molecular & Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| |
Collapse
|
19
|
Long AM, Kwon JM, Lee G, Reiser NL, Vaught LA, O'Brien JG, Page PGT, Hadhazy M, Reynolds JC, Crosbie RH, Demonbreun AR, McNally EM. The extracellular matrix differentially directs myoblast motility and differentiation in distinct forms of muscular dystrophy: Dystrophic matrices alter myoblast motility. Matrix Biol 2024; 129:44-58. [PMID: 38582404 PMCID: PMC11104166 DOI: 10.1016/j.matbio.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024]
Abstract
Extracellular matrix (ECM) pathologic remodeling underlies many disorders, including muscular dystrophy. Tissue decellularization removes cellular components while leaving behind ECM components. We generated "on-slide" decellularized tissue slices from genetically distinct dystrophic mouse models. The ECM of dystrophin- and sarcoglycan-deficient muscles had marked thrombospondin 4 deposition, while dysferlin-deficient muscle had excess decorin. Annexins A2 and A6 were present on all dystrophic decellularized ECMs, but annexin matrix deposition was excessive in dysferlin-deficient muscular dystrophy. Muscle-directed viral expression of annexin A6 resulted in annexin A6 in the ECM. C2C12 myoblasts seeded onto decellularized matrices displayed differential myoblast mobility and fusion. Dystrophin-deficient decellularized matrices inhibited myoblast mobility, while dysferlin-deficient decellularized matrices enhanced myoblast movement and differentiation. Myoblasts treated with recombinant annexin A6 increased mobility and fusion like that seen on dysferlin-deficient decellularized matrix and demonstrated upregulation of ECM and muscle cell differentiation genes. These findings demonstrate specific fibrotic signatures elicit effects on myoblast activity.
Collapse
Affiliation(s)
- Ashlee M Long
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jason M Kwon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - GaHyun Lee
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nina L Reiser
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lauren A Vaught
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph G O'Brien
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Patrick G T Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA; Department of Neurology David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA; Department of Neurology David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
20
|
Kristensen MA, Rich KK, Mogensen TC, Damsgaard Jensen AM, Fex Svenningsen Å, Zhang M. Focal Traumatic Brain Injury Impairs the Integrity of the Basement Membrane of Hindlimb Muscle Fibers Revealed by Extracellular Matrix Immunoreactivity. Life (Basel) 2024; 14:543. [PMID: 38792565 PMCID: PMC11121831 DOI: 10.3390/life14050543] [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: 02/12/2024] [Revised: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Traumatic brain injury (TBI) stands as a prominent global cause of disability, with motor deficits being a common consequence. Despite its widespread impact, the precise pathological mechanisms underlying motor deficits after TBI remain elusive. In this study, hindlimb postural asymmetry (HL-PA) development in rats subjected to focal TBI was investigated to explore the potential roles of collagen IV and laminin within the extracellular matrix (ECM) of selected hindlimb muscles in the emergence of motor deficits following TBI. A focal TBI was induced by ablating the left sensorimotor cortex in rats and motor deficits were assessed by measuring HL-PA. The expression of laminin and collagen IV in eight selected muscles on each side of the hindlimbs from both TBI- and sham-operated rats were studied using immunohistochemistry and semi-quantitatively analyzed. The results indicated that the TBI rats exhibited HL-PA, characterized by flexion of the contralateral (right) hindlimb. In the sham-operated rats, the immunoreactive components of laminin and collagen IV were evenly and smoothly distributed along the border of the muscle fibers in all the investigated muscles. In contrast, in the TBI rats, the pattern was broken into aggregated, granule-like, immunoreactive components. Such a labeling pattern was detected in all the investigated muscles both from the contra- and ipsilateral sides of the TBI rats. However, in TBI rats, most of the muscles from the contralateral hindlimb showed a significantly increased expression of these two proteins in comparison with those from the ipsilateral hindlimb. In comparison to sham-operated rats, there was a significant increase in laminin and collagen IV expression in various contralateral hindlimb muscles in the TBI rats. These findings suggest potential implications of laminin and collagen IV in the development of motor deficits following a focal TBI.
Collapse
Affiliation(s)
- Mette Albæk Kristensen
- Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense, Denmark; (M.A.K.); (K.K.R.); (T.C.M.); (Å.F.S.)
| | - Karen Kalhøj Rich
- Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense, Denmark; (M.A.K.); (K.K.R.); (T.C.M.); (Å.F.S.)
| | - Tobias Christian Mogensen
- Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense, Denmark; (M.A.K.); (K.K.R.); (T.C.M.); (Å.F.S.)
| | | | - Åsa Fex Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense, Denmark; (M.A.K.); (K.K.R.); (T.C.M.); (Å.F.S.)
- Brain Research—Inter Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, DK-5230 Odense, Denmark
| | - Mengliang Zhang
- Department of Molecular Medicine, University of Southern Denmark, DK-5230 Odense, Denmark; (M.A.K.); (K.K.R.); (T.C.M.); (Å.F.S.)
- Brain Research—Inter Disciplinary Guided Excellence (BRIDGE), University of Southern Denmark, DK-5230 Odense, Denmark
| |
Collapse
|
21
|
Guo Q, Luo Q, Song G. Control of muscle satellite cell function by specific exercise-induced cytokines and their applications in muscle maintenance. J Cachexia Sarcopenia Muscle 2024; 15:466-476. [PMID: 38375571 PMCID: PMC10995279 DOI: 10.1002/jcsm.13440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 02/21/2024] Open
Abstract
Exercise is recognized to play an observable role in improving human health, especially in promoting muscle hypertrophy and intervening in muscle mass loss-related diseases, including sarcopenia. Recent rapid advances have demonstrated that exercise induces the release of abundant cytokines from several tissues (e.g., liver, muscle, and adipose tissue), and multiple cytokines improve the functions or expand the numbers of adult stem cells, providing candidate cytokines for alleviating a wide range of diseases. Muscle satellite cells (SCs) are a population of muscle stem cells that are mitotically quiescent but exit from the dormancy state to become activated in response to physical stimuli, after which SCs undergo asymmetric divisions to generate new SCs (stem cell pool maintenance) and commit to later differentiation into myocytes (skeletal muscle replenishment). SCs are essential for the postnatal growth, maintenance, and regeneration of skeletal muscle. Emerging evidence reveals that exercise regulates muscle function largely via the exercise-induced cytokines that govern SC potential, but this phenomenon is complicated and confusing. This review provides a comprehensive integrative overview of the identified exercise-induced cytokines and the roles of these cytokines in SC function, providing a more complete picture regarding the mechanism of SC homeostasis and rejuvenation therapies for skeletal muscle.
Collapse
Affiliation(s)
- Qian Guo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqingChina
| |
Collapse
|
22
|
Olson LC, Nguyen T, Sabalewski EL, Puetzer JL, Schwartz Z, McClure MJ. S100b treatment overcomes RAGE signaling deficits in myoblasts on advanced glycation end-product cross-linked collagen and promotes myogenic differentiation. Am J Physiol Cell Physiol 2024; 326:C1080-C1093. [PMID: 38314727 DOI: 10.1152/ajpcell.00502.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Advanced glycation end-products (AGEs) stochastically accrue in skeletal muscle and on collagen over an individual's lifespan, stiffening the muscle and modifying the stem cell (MuSC) microenvironment while promoting proinflammatory, antiregenerative signaling via the receptor for advanced glycation end-products (RAGEs). In the present study, a novel in vitro model was developed of this phenomenon by cross linking a 3-D collagen scaffold with AGEs and investigating how myoblasts responded to such an environment. Briefly, collagen scaffolds were incubated with d-ribose (0, 25, 40, 100, or 250 mM) for 5 days at 37°C. C2C12 immortalized mouse myoblasts were grown on the scaffolds for 6 days in growth conditions for proliferation, and 12 days for differentiation and fusion. Human primary myoblasts were also used to confirm the C2C12 data. AGEs aberrantly extended the DNA production stage of C2C12s (but not in human primary myoblasts) which is known to delay differentiation in myogenesis, and this effect was prevented by RAGE inhibition. Furthermore, the differentiation and fusion of myoblasts were disrupted by AGEs, which were associated with reductions in integrins and suppression of RAGE. The addition of S100b (RAGE agonist) recovered the differentiation and fusion of myoblasts, and the addition of RAGE inhibitors (FPS-ZM1 and Azeliragon) inhibited the differentiation and fusion of myoblasts. Our results provide novel insights into the role of the AGE-RAGE axis in skeletal muscle aging, and future work is warranted on the potential application of S100b as a proregenerative factor in aged skeletal muscle.NEW & NOTEWORTHY Collagen cross-linked by advanced glycation end-products (AGEs) induced myoblast proliferation but prevented differentiation, myotube formation, and RAGE upregulation. RAGE inhibition occluded AGE-induced myoblast proliferation, while the delivery of S100b, a RAGE ligand, recovered fusion deficits.
Collapse
Affiliation(s)
- Lucas C Olson
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Gerontology, College of Health Professionals, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Tri Nguyen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Eleanor L Sabalewski
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Orthopaedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Michael J McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Orthopaedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
23
|
Nguyen J, Wang L, Lei W, Hu Y, Gulati N, Chavez-Madero C, Ahn H, Ginsberg HJ, Krawetz R, Brandt M, Betz T, Gilbert PM. Culture substrate stiffness impacts human myoblast contractility-dependent proliferation and nuclear envelope wrinkling. J Cell Sci 2024; 137:jcs261666. [PMID: 38345101 PMCID: PMC11033523 DOI: 10.1242/jcs.261666] [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: 09/27/2023] [Accepted: 02/04/2024] [Indexed: 03/28/2024] Open
Abstract
Understanding how biophysical and biochemical microenvironmental cues together influence the regenerative activities of muscle stem cells and their progeny is crucial in strategizing remedies for pathological dysregulation of these cues in aging and disease. In this study, we investigated the cell-level influences of extracellular matrix (ECM) ligands and culture substrate stiffness on primary human myoblast contractility and proliferation within 16 h of plating and found that tethered fibronectin led to stronger stiffness-dependent responses compared to laminin and collagen. A proteome-wide analysis further uncovered cell metabolism, cytoskeletal and nuclear component regulation distinctions between cells cultured on soft and stiff substrates. Interestingly, we found that softer substrates increased the incidence of myoblasts with a wrinkled nucleus, and that the extent of wrinkling could predict Ki67 (also known as MKI67) expression. Nuclear wrinkling and Ki67 expression could be controlled by pharmacological manipulation of cellular contractility, offering a potential cellular mechanism. These results provide new insights into the regulation of human myoblast stiffness-dependent contractility response by ECM ligands and highlight a link between myoblast contractility and proliferation.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Lu Wang
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Wen Lei
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Yechen Hu
- Department of Chemistry, University of Toronto, Toronto, ON, M5S 3H6, Canada
| | - Nitya Gulati
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Carolina Chavez-Madero
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Henry Ahn
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
| | - Howard J. Ginsberg
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2C4, Canada
- Li Ka Shing Knowledge Institute, Saint Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Roman Krawetz
- McCaig Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Matthias Brandt
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University Münster, 48149 Münster, Germany
| | - Timo Betz
- Third Institute of Physics – Biophysics, Georg August University Göttingen, 37077 Göttingen, Germany
| | - Penney M. Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3E2, Canada
- Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5, Canada
| |
Collapse
|
24
|
Helzer D, Kannan P, Reynolds JC, Gibbs DE, Crosbie RH. Role of microenvironment on muscle stem cell function in health, adaptation, and disease. Curr Top Dev Biol 2024; 158:179-201. [PMID: 38670705 DOI: 10.1016/bs.ctdb.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The role of the cellular microenvironment has recently gained attention in the context of muscle health, adaption, and disease. Emerging evidence supports major roles for the extracellular matrix (ECM) in regeneration and the dynamic regulation of the satellite cell niche. Satellite cells normally reside in a quiescent state in healthy muscle, but upon muscle injury, they activate, proliferate, and fuse to the damaged fibers to restore muscle function and architecture. This chapter reviews the composition and mechanical properties of skeletal muscle ECM and the role of these factors in contributing to the satellite cell niche that impact muscle regeneration. In addition, the chapter details the effects of satellite cell-matrix interactions and provides evidence that there is bidirectional regulation affecting both the cellular and extracellular microenvironment within skeletal muscle. Lastly, emerging methods to investigate satellite cell-matrix interactions will be presented.
Collapse
Affiliation(s)
- Daniel Helzer
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Pranav Kannan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Devin E Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, 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.
| |
Collapse
|
25
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
26
|
Chrysostomou E, Mourikis P. The extracellular matrix niche of muscle stem cells. Curr Top Dev Biol 2024; 158:123-150. [PMID: 38670702 DOI: 10.1016/bs.ctdb.2024.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Preserving the potency of stem cells in adult tissues is very demanding and relies on the concerted action of various cellular and non-cellular elements in a precise stoichiometry. This balanced microenvironment is found in specific anatomical "pockets" within the tissue, known as the stem cell niche. In this review, we explore the interplay between stem cells and their niches, with a primary focus on skeletal muscle stem cells and the extracellular matrix (ECM). Quiescent muscle stem cells, known as satellite cells are active producers of a diverse array of ECM molecules, encompassing major constituents like collagens, laminins, and integrins, some of which are explored in this review. The conventional perception of ECM as merely a structural scaffold is evolving. Collagens can directly interact as ligands with receptors on satellite cells, while other ECM proteins have the capacity to sequester growth factors and regulate their release, especially relevant during satellite cell turnover in homeostasis or activation upon injury. Additionally, we explore an evolutionary perspective on the ECM across a range of multicellular organisms and discuss a model wherein satellite cells are self-sustained by generating their own niche. Considering the prevalence of ECM proteins in the connective tissue of various organs it is not surprising that mutations in ECM genes have pathological implications, including in muscle, where they can lead to myopathies. However, the particular role of certain disease-related ECM proteins in stem cell maintenance highlights the potential contribution of stem cell deregulation to the progression of these disorders.
Collapse
Affiliation(s)
- Eleni Chrysostomou
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), Créteil, France
| | - Philippos Mourikis
- Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale (INSERM), Mondor Institute for Biomedical Research (IMRB), Créteil, France.
| |
Collapse
|
27
|
Girolamo DD, Benavente-Diaz M, Murolo M, Grimaldi A, Lopes PT, Evano B, Kuriki M, Gioftsidi S, Laville V, Tinevez JY, Letort G, Mella S, Tajbakhsh S, Comai G. Extraocular muscle stem cells exhibit distinct cellular properties associated with non-muscle molecular signatures. Development 2024; 151:dev202144. [PMID: 38240380 DOI: 10.1242/dev.202144] [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: 06/29/2023] [Accepted: 12/27/2023] [Indexed: 02/22/2024]
Abstract
Skeletal muscle stem cells (MuSCs) are recognised as functionally heterogeneous. Cranial MuSCs are reported to have greater proliferative and regenerative capacity when compared with those in the limb. A comprehensive understanding of the mechanisms underlying this functional heterogeneity is lacking. Here, we have used clonal analysis, live imaging and single cell transcriptomic analysis to identify crucial features that distinguish extraocular muscle (EOM) from limb muscle stem cell populations. A MyogeninntdTom reporter showed that the increased proliferation capacity of EOM MuSCs correlates with deferred differentiation and lower expression of the myogenic commitment gene Myod. Unexpectedly, EOM MuSCs activated in vitro expressed a large array of extracellular matrix components typical of mesenchymal non-muscle cells. Computational analysis underscored a distinct co-regulatory module, which is absent in limb MuSCs, as driver of these features. The EOM transcription factor network, with Foxc1 as key player, appears to be hardwired to EOM identity as it persists during growth, disease and in vitro after several passages. Our findings shed light on how high-performing MuSCs regulate myogenic commitment by remodelling their local environment and adopting properties not generally associated with myogenic cells.
Collapse
Affiliation(s)
- Daniela Di Girolamo
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Maria Benavente-Diaz
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005 Paris, France
| | - Melania Murolo
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Alexandre Grimaldi
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005 Paris, France
| | - Priscilla Thomas Lopes
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Brendan Evano
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Mao Kuriki
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Stamatia Gioftsidi
- Université Paris-Est, 77420 Champs-sur- Marne, France
- Freie Universität Berlin, 14195 Berlin, Germany
- Inserm, IMRB U955-E10, 94000 Créteil, France
| | - Vincent Laville
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Jean-Yves Tinevez
- Institut Pasteur, Université Paris Cité, Image Analysis Hub, 75015 Paris, France
| | - Gaëlle Letort
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Sebastian Mella
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Glenda Comai
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| |
Collapse
|
28
|
Metti S, Da Ros F, Toniato G, Cescon M, Bonaldo P. Native collagen VI delays early muscle stem cell differentiation. J Cell Sci 2024; 137:jcs261419. [PMID: 38224152 PMCID: PMC10911284 DOI: 10.1242/jcs.261419] [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: 06/17/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024] Open
Abstract
Adult muscle stem cells (MuSCs) are critical for muscle homeostasis and regeneration, and their behavior relies on a finely regulated niche made of specific extracellular matrix (ECM) components and soluble factors. Among ECM proteins, collagen VI (Col6) influences the mechanical properties of the niche and, in turn, MuSC self-renewal capabilities. Here, we investigated whether Col6 can exert a direct function as a biochemical signal for regulating the stemness and differentiation of murine MuSCs and myoblasts. Native Col6, but not its pepsin-resistant fragment, counteracts the early differentiation of myogenic cells by reducing the expression of differentiation marker genes and preserving stemness features, with inhibition of the canonical Wnt pathway. Our data indicate that extracellular Col6 acts as a soluble ligand in delaying early myogenic differentiation by regulating intracellular signals involved in adult myogenesis.
Collapse
Affiliation(s)
- Samuele Metti
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Francesco Da Ros
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Giorgia Toniato
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| |
Collapse
|
29
|
Jiang H, Liu B, Lin J, Xue T, Han Y, Lu C, Zhou S, Gu Y, Xu F, Shen Y, Xu L, Sun H. MuSCs and IPCs: roles in skeletal muscle homeostasis, aging and injury. Cell Mol Life Sci 2024; 81:67. [PMID: 38289345 PMCID: PMC10828015 DOI: 10.1007/s00018-023-05096-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Skeletal muscle is a highly specialized tissue composed of myofibres that performs crucial functions in movement and metabolism. In response to external stimuli and injuries, a range of stem/progenitor cells, with muscle stem cells or satellite cells (MuSCs) being the predominant cell type, are rapidly activated to repair and regenerate skeletal muscle within weeks. Under normal conditions, MuSCs remain in a quiescent state, but become proliferative and differentiate into new myofibres in response to injury. In addition to MuSCs, some interstitial progenitor cells (IPCs) such as fibro-adipogenic progenitors (FAPs), pericytes, interstitial stem cells expressing PW1 and negative for Pax7 (PICs), muscle side population cells (SPCs), CD133-positive cells and Twist2-positive cells have been identified as playing direct or indirect roles in regenerating muscle tissue. Here, we highlight the heterogeneity, molecular markers, and functional properties of these interstitial progenitor cells, and explore the role of muscle stem/progenitor cells in skeletal muscle homeostasis, aging, and muscle-related diseases. This review provides critical insights for future stem cell therapies aimed at treating muscle-related diseases.
Collapse
Affiliation(s)
- Haiyan Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tong Xue
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Yimin Han
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Lingchi Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
30
|
Duran P, Yang BA, Plaster E, Eiken M, Loebel C, Aguilar CA. Quantification of local matrix deposition during muscle stem cell activation using engineered hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576326. [PMID: 38328131 PMCID: PMC10849481 DOI: 10.1101/2024.01.20.576326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Adult stem cells occupy a niche that contributes to their function, but how stem cells remodel their microenvironment remains an open-ended question. Herein, biomaterials-based systems and metabolic labeling were utilized to evaluate how skeletal muscle stem cells deposit extracellular matrix. Muscle stem cells and committed myoblasts were observed to generate less nascent matrix than muscle resident fibro-adipogenic progenitors. When cultured on substrates that matched the stiffness of physiological uninjured and injured muscles, the increased nascent matrix deposition was associated with stem cell activation. Reducing the ability to deposit nascent matrix in muscle stem cells attenuated function and mimicked impairments observed from muscle stem cells isolated from old aged muscles, which could be rescued with therapeutic supplementation of insulin-like growth factors. These results highlight how nascent matrix production is critical for maintaining healthy stem cell function.
Collapse
Affiliation(s)
- Pamela Duran
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Benjamin A. Yang
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eleanor Plaster
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Madeline Eiken
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Claudia Loebel
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Dept. of Materials Science & Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carlos A. Aguilar
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- BioInterfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
31
|
Wang Q, Liang Q, Dou J, Zhou H, Zeng C, Pan H, Shen Y, Li Q, Liu Y, Leong DT, Jiang W, Wang Y. Breaking through the basement membrane barrier to improve nanotherapeutic delivery to tumours. NATURE NANOTECHNOLOGY 2024; 19:95-105. [PMID: 37709950 DOI: 10.1038/s41565-023-01498-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/03/2023] [Indexed: 09/16/2023]
Abstract
An effective nanotherapeutic transport from the vasculature to the tumour is crucial for cancer treatment with minimal side effects. Here we demonstrate that, in addition to the endothelial barrier, the tumour vascular basement membrane surrounding the endothelium acts as a formidable mechanical barrier that entraps nanoparticles (NPs) in the subendothelial void, forming perivascular NP pools. Breaking through this basement membrane barrier substantially increases NP extravasation. Using inflammation triggered by local hyperthermia, we develop a cooperative immunodriven strategy to overcome the basement membrane barrier that leads to robust tumour killing. Hyperthermia-triggered accumulation and inflammation of platelets attract neutrophils to the NP pools. The subsequent movement of neutrophils through the basement membrane can release the NPs entrapped in the subendothelial void, resulting in increased NP penetration into deeper tumours. We show the necessity of considering the tumour vascular basement membrane barrier when delivering nanotherapeutics. Understanding this barrier will contribute to developing more effective antitumour therapies.
Collapse
Affiliation(s)
- Qin Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qirui Liang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaxiang Dou
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Han Zhou
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cici Zeng
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huimin Pan
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanqiong Shen
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Quan Li
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yi Liu
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
| | - Wei Jiang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Yucai Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
| |
Collapse
|
32
|
Grobbelaar S, Mercier AE, van den Bout I, Durandt C, Pepper MS. Considerations for enhanced mesenchymal stromal/stem cell myogenic commitment in vitro. Clin Transl Sci 2024; 17:e13703. [PMID: 38098144 PMCID: PMC10787211 DOI: 10.1111/cts.13703] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/16/2023] [Accepted: 12/09/2023] [Indexed: 01/15/2024] Open
Abstract
The generation of tissue from stem cells is an alluring concept as it holds a number of potential applications in clinical therapeutics and regenerative medicine. Mesenchymal stromal/stem cells (MSCs) can be isolated from a number of different somatic sources, and have the capacity to differentiate into adipogenic, osteogenic, chondrogenic, and myogenic lineages. Although the first three have been extensively investigated, there remains a paucity of literature on the latter. This review looks at the various strategies available in vitro to enhance harvested MSC commitment and differentiation into the myogenic pathway. These include chemical inducers, myogenic-enhancing cell culture substrates, and mechanical and dynamic culturing conditions. Drawing on information from embryonic and postnatal myogenesis from somites, satellite, and myogenic progenitor cells, the mechanisms behind the chemical and mechanical induction strategies can be studied, and the sequential gene and signaling cascades can be used to monitor the progression of myogenic differentiation in the laboratory. Increased understanding of the stimuli and signaling mechanisms in the initial stages of MSC myogenic commitment will provide tools with which we can enhance their differentiation efficacy and advance the process to clinical translation.
Collapse
Affiliation(s)
- Simone Grobbelaar
- Department of Physiology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Anne E. Mercier
- Department of Physiology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Iman van den Bout
- Department of Physiology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
- Centre for Neuroendocrinology, Department of Immunology, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| |
Collapse
|
33
|
Qiu X, Wang HY, Yang ZY, Sun LM, Liu SN, Fan CQ, Zhu F. Uncovering the prominent role of satellite cells in paravertebral muscle development and aging by single-nucleus RNA sequencing. Genes Dis 2023; 10:2597-2613. [PMID: 37554180 PMCID: PMC10404979 DOI: 10.1016/j.gendis.2023.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/06/2022] [Accepted: 01/02/2023] [Indexed: 02/05/2023] Open
Abstract
To uncover the role of satellite cells (SCs) in paravertebral muscle development and aging, we constructed a single-nucleus transcriptomic atlas of mouse paravertebral muscle across seven timepoints spanning the embryo (day 16.5) to old (month 24) stages. Eight cell types, including SCs, fast muscle cells, and slow muscle cells, were identified. An energy metabolism-related gene set, TCA CYCLE IN SENESCENCE, was enriched in SCs. Forty-two skeletal muscle disease-related genes were highly expressed in SCs and exhibited similar expression patterns. Among them, Pdha1 was the core gene in the TCA CYCLE IN SENESCENCE; Pgam2, Sod1, and Suclg1 are transcription factors closely associated with skeletal muscle energy metabolism. Transcription factor enrichment analysis of the 42 genes revealed that Myod1 and Mef2a were also highly expressed in SCs, which regulated Pdha1 expression and were associated with skeletal muscle development. These findings hint that energy metabolism may be pivotal in SCs development and aging. Three ligand-receptor pairs of extracellular matrix (ECM)-receptor interactions, Lamc1-Dag1, Lama2-Dag1, and Hspg2-Dag1, may play a vital role in SCs interactions with slow/fast muscle cells and SCs self-renewal. Finally, we built the first database of a skeletal muscle single-cell transcriptome, the Musculoskeletal Cell Atlas (http://www.mskca.tech), which lists 630,040 skeletal muscle cells and provides interactive visualization, a useful resource for revealing skeletal muscle cellular heterogeneity during development and aging. Our study could provide new targets and ideas for developing drugs to inhibit skeletal muscle aging and treat skeletal muscle diseases.
Collapse
Affiliation(s)
- Xin Qiu
- Department of Spinal Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong 518000, China
- Department of Orthopedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Hao-Yu Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100000, China
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI-Qingdao, Qingdao, Shandong 266000, China
| | - Zhen-Yu Yang
- Department of Spinal Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong 518000, China
| | - Li-Ming Sun
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Forth Military Medical University, Xi'an, Shaanxi 710000, China
| | - Shu-Nan Liu
- Department of Spinal Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong 518000, China
| | - Chui-Qin Fan
- China Medical University, Shenyang, Liaoning 110000, China
| | - Feng Zhu
- Department of Spinal Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong 518000, China
- Department of Orthopedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
34
|
Han S, Cruz SH, Park S, Shin SR. Nano-biomaterials and advanced fabrication techniques for engineering skeletal muscle tissue constructs in regenerative medicine. NANO CONVERGENCE 2023; 10:48. [PMID: 37864632 PMCID: PMC10590364 DOI: 10.1186/s40580-023-00398-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023]
Abstract
Engineered three-dimensional (3D) tissue constructs have emerged as a promising solution for regenerating damaged muscle tissue resulting from traumatic or surgical events. 3D architecture and function of the muscle tissue constructs can be customized by selecting types of biomaterials and cells that can be engineered with desired shapes and sizes through various nano- and micro-fabrication techniques. Despite significant progress in this field, further research is needed to improve, in terms of biomaterials properties and fabrication techniques, the resemblance of function and complex architecture of engineered constructs to native muscle tissues, potentially enhancing muscle tissue regeneration and restoring muscle function. In this review, we discuss the latest trends in using nano-biomaterials and advanced nano-/micro-fabrication techniques for creating 3D muscle tissue constructs and their regeneration ability. Current challenges and potential solutions are highlighted, and we discuss the implications and opportunities of a future perspective in the field, including the possibility for creating personalized and biomanufacturable platforms.
Collapse
Affiliation(s)
- Seokgyu Han
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Sebastián Herrera Cruz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA.
| |
Collapse
|
35
|
Szabó L, Telek A, Fodor J, Dobrosi N, Dócs K, Hegyi Z, Gönczi M, Csernoch L, Dienes B. Reduced Expression of Septin7 Hinders Skeletal Muscle Regeneration. Int J Mol Sci 2023; 24:13536. [PMID: 37686339 PMCID: PMC10487768 DOI: 10.3390/ijms241713536] [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: 06/05/2023] [Revised: 07/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Septins are considered the fourth component of the cytoskeleton with the septin7 isoform playing a critical role in the formation of diffusion barriers in phospholipid bilayers and intra- and extracellular scaffolds. While its importance has already been confirmed in different intracellular processes, very little is known about its role in skeletal muscle. Muscle regeneration was studied in a Sept7 conditional knock-down mouse model to prove the possible role of septin7 in this process. Sterile inflammation in skeletal muscle was induced which was followed by regeneration resulting in the upregulation of septin7 expression. Partial knock-down of Sept7 resulted in an increased number of inflammatory cells and myofibers containing central nuclei. Taken together, our data suggest that partial knock-down of Sept7 hinders the kinetics of muscle regeneration, indicating its crucial role in skeletal muscle functions.
Collapse
Affiliation(s)
- László Szabó
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Andrea Telek
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Nóra Dobrosi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Klaudia Dócs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Zoltán Hegyi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mónika Gönczi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, University of Debrecen, 4032 Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
36
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [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] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
37
|
Fujita R, Mizuno S, Sadahiro T, Hayashi T, Sugasawa T, Sugiyama F, Ono Y, Takahashi S, Ieda M. Generation of a MyoD knock-in reporter mouse line to study muscle stem cell dynamics and heterogeneity. iScience 2023; 26:106592. [PMID: 37250337 PMCID: PMC10214404 DOI: 10.1016/j.isci.2023.106592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/19/2023] [Accepted: 03/31/2023] [Indexed: 05/31/2023] Open
Abstract
Myoblast determination protein 1 (MyoD) dynamics define the activation status of muscle stem cells (MuSCs), aiding in muscle tissue regeneration after injury. However, the lack of experimental platforms to monitor MyoD dynamics in vitro and in vivo has hampered the investigation of fate determination and heterogeneity of MuSCs. Herein, we report a MyoD knock-in (MyoD-KI) reporter mouse expressing tdTomato at the endogenous MyoD locus. Expression of tdTomato in MyoD-KI mice recapitulated the endogenous MyoD expression dynamics in vitro and during the early phase of regeneration in vivo. Additionally, we showed that tdTomato fluorescence intensity defines MuSC activation status without immunostaining. Based on these features, we developed a high-throughput screening system to assess the effects of drugs on the behavior of MuSCs in vitro. Thus, MyoD-KI mice are an invaluable resource for studying the dynamics of MuSCs, including their fate decisions and heterogeneity, and for drug screening in stem cell therapy.
Collapse
Affiliation(s)
- Ryo Fujita
- Division of Regenerative Medicine, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Taketaro Sadahiro
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takuto Hayashi
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Takehito Sugasawa
- Laboratory of Clinical Examination and Sports Medicine, Department of Clinical Medicine, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masaki Ieda
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
38
|
Bobadilla Muñoz M, Orbe J, Abizanda G, Machado FJD, Vilas A, Ullate-Agote A, Extramiana L, Baraibar Churio A, Aranguren XL, Cantero G, Sáinz Amillo N, Rodríguez JA, Ramos García L, Romero Riojas JP, Vallejo-Illarramendi A, Paradas C, López de Munain A, Páramo JA, Prósper F, Pérez-Ruiz A. Loss of the matrix metalloproteinase-10 causes premature features of aging in satellite cells. Front Cell Dev Biol 2023; 11:1128534. [PMID: 37228645 PMCID: PMC10203875 DOI: 10.3389/fcell.2023.1128534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Aged muscles accumulate satellite cells with a striking decline response to damage. Although intrinsic defects in satellite cells themselves are the major contributors to aging-associated stem cell dysfunction, increasing evidence suggests that changes in the muscle-stem cell local microenvironment also contribute to aging. Here, we demonstrate that loss of the matrix metalloproteinase-10 (MMP-10) in young mice alters the composition of the muscle extracellular matrix (ECM), and specifically disrupts the extracellular matrix of the satellite cell niche. This situation causes premature features of aging in the satellite cells, contributing to their functional decline and a predisposition to enter senescence under proliferative pressure. Similarly, reduction of MMP-10 levels in young satellite cells from wild type animals induces a senescence response, while addition of the protease delays this program. Significantly, the effect of MMP-10 on satellite cell aging can be extended to another context of muscle wasting, muscular dystrophy. Systemic treatment of mdx dystrophic mice with MMP-10 prevents the muscle deterioration phenotype and reduces cellular damage in the satellite cells, which are normally under replicative pressure. Most importantly, MMP-10 conserves its protective effect in the satellite cell-derived myoblasts isolated from a Duchenne muscular dystrophy patient by decreasing the accumulation of damaged DNA. Hence, MMP-10 provides a previously unrecognized therapeutic opportunity to delay satellite cell aging and overcome satellite cell dysfunction in dystrophic muscles.
Collapse
Affiliation(s)
- Miriam Bobadilla Muñoz
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Josune Orbe
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS)-Ictus, Instituto de Salud Carlos III, Madrid, Spain
| | - Gloria Abizanda
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Florencio J. D. Machado
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
| | - Amaia Vilas
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Asier Ullate-Agote
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leire Extramiana
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Arantxa Baraibar Churio
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier L. Aranguren
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gloria Cantero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Neuromuscular Disorders Unit, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Neira Sáinz Amillo
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Centre for Nutrition Research, Universidad de Navarra, Pamplona, Spain
| | - José Antonio Rodríguez
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Ramos García
- Radiology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Radiology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, San Sebastian, Spain
| | - Juan Pablo Romero Riojas
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | - Carmen Paradas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Neuromuscular Disorders Unit, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Adolfo López de Munain
- CIBERNED-Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
- Neurology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, San Sebastian, Spain
| | - José Antonio Páramo
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Service, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
| | - Ana Pérez-Ruiz
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
39
|
Singh AK, Peng BY, Chien ST, Chan CH, Deng YH, Pai HY, Wei HJ, Wang MF, Wang SH, Wu CY, Deng WP. Anti-aging biomaterial sturgeon chondroitin sulfate upregulating anti-oxidant and SIRT-1/c-fos gene expression to reprogram stem cell senescence and prolong longevity. Biomater Sci 2023. [PMID: 37158091 DOI: 10.1039/d2bm01997c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Aging involves tissue and cell potential dysfunction characterized by stem cell senescence and extracellular matrix microenvironment (ECM) alteration. Chondroitin sulfate (CS), found in the ECM of normal cells and tissues, aids in maintaining tissue homeostasis. Here, CS-derived biomaterial (CSDB) from sturgeon is extracted to investigate its antiaging effect in senescence-accelerated mouse prone-8 (SAMP8) mice and elucidate the underlying mechanism of its action. Although CSDB has been widely extracted from different sources and used as a scaffold, hydrogel, or drug carrier for the treatment of various pathological diseases, CSDB has not yet been used as a biomaterial for the amelioration of senescence and aging features. In this study, the extracted sturgeon CSDB showed a low molecular weight and comprised 59% 4-sulfated CS and 23% 6-sulfated CS. In an in vitro study, sturgeon CSDB promoted cell proliferation and reduced oxidative stress to inhibit stem cell senescence. In an ex vivo study, after oral CSDB treatment of SAMP8 mice, the stem cells were extracted to analyze the p16Ink4a and p19Arf gene-related pathways, which were inhibited and then SIRT-1 gene expression was upregulated to reprogram stem cells from a senescence state for retarding aging. In an in vivo study, CSDB also restored the aging-phenotype-related bone mineral density and skin morphology to prolong longevity. Thus, sturgeon CSDB may be useful for prolonging healthy longevity as an anti-aging drug.
Collapse
Affiliation(s)
- Abhinay Kumar Singh
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Bou-Yue Peng
- Department of Dentistry, Taipei Medical University Hospital, Taipei 110301, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Shaw-Ting Chien
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Chun-Hao Chan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Yue-Hua Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Hsiao-Yu Pai
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110301, Taiwan
| | - Hong-Jian Wei
- Department of Radiation Oncology, Columbia University Irving Medical Center, 630 W. 168th Street, New York, NY, 10032, USA
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University, Taichung 433303, Taiwan
| | - Shwu-Huey Wang
- Core Facility Center, Department of Research Development, Taipei Medical University, Taipei 11030, Taiwan
| | - Chia-Yu Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei 110301, Taiwan.
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Taipei 242062, Taiwan
| |
Collapse
|
40
|
Aleksandrowicz R, Strączkowski M. Link between insulin resistance and skeletal muscle extracellular matrix remodeling. Endocr Connect 2023; 12:e230023. [PMID: 36917038 PMCID: PMC10160556 DOI: 10.1530/ec-23-0023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/16/2023]
Abstract
Skeletal muscle is the main metabolic tissue responsible for glucose homeostasis in the body. It is surrounded by the extracellular matrix (ECM) consisting of three layers: epimysium, perimysium, and endomysium. ECM plays an important role in the muscle, as it provides integrity and scaffolding cells. The observed disturbances in this structure are related to the abnormal remodeling of the ECM (through an increase in the concentration of its components). ECM rearrangement may impair insulin action by increasing the physical barrier to insulin transport and reducing insulin transport into muscle cells as well as by directly inhibiting insulin action through integrin signaling. Thus, improper ECM remodeling may contribute to the development of insulin resistance (IR) and related comorbidities. In turn, IR-associated conditions may further aggravate disturbances of ECM in skeletal muscle. This review describes the major components of the ECM that are necessary for its proper function. Particular attention was also paid to receptors (integrins) involved in the signaling of metabolic pathways. Finally, changes in ECM components in the context of clinical and animal studies are discussed. This article will help the reader to systematize knowledge related to the ECM and to better understand the relationship between ECM remodeling and IR, and its role in the pathogenesis of T2DM. The information in this article presents the concept of the role of ECM and its remodeling in the pathogenesis of IR, which may contribute to developing new therapeutic solutions.
Collapse
Affiliation(s)
- Róża Aleksandrowicz
- Department of Prophylaxis of Metabolic Diseases, Bialystok, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Marek Strączkowski
- Department of Prophylaxis of Metabolic Diseases, Bialystok, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
41
|
Stearns-Reider KM, Hicks MR, Hammond KG, Reynolds JC, Maity A, Kurmangaliyev YZ, Chin J, Stieg AZ, Geisse NA, Hohlbauch S, Kaemmer S, Schmitt LR, Pham TT, Yamauchi K, Novitch BG, Wollman R, Hansen KC, Pyle AD, Crosbie RH. Myoscaffolds reveal laminin scarring is detrimental for stem cell function while sarcospan induces compensatory fibrosis. NPJ Regen Med 2023; 8:16. [PMID: 36922514 PMCID: PMC10017766 DOI: 10.1038/s41536-023-00287-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
We developed an on-slide decellularization approach to generate acellular extracellular matrix (ECM) myoscaffolds that can be repopulated with various cell types to interrogate cell-ECM interactions. Using this platform, we investigated whether fibrotic ECM scarring affected human skeletal muscle progenitor cell (SMPC) functions that are essential for myoregeneration. SMPCs exhibited robust adhesion, motility, and differentiation on healthy muscle-derived myoscaffolds. All SPMC interactions with fibrotic myoscaffolds from dystrophic muscle were severely blunted including reduced motility rate and migration. Furthermore, SMPCs were unable to remodel laminin dense fibrotic scars within diseased myoscaffolds. Proteomics and structural analysis revealed that excessive collagen deposition alone is not pathological, and can be compensatory, as revealed by overexpression of sarcospan and its associated ECM receptors in dystrophic muscle. Our in vivo data also supported that ECM remodeling is important for SMPC engraftment and that fibrotic scars may represent one barrier to efficient cell therapy.
Collapse
Affiliation(s)
- Kristen M Stearns-Reider
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael R Hicks
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Katherine G Hammond
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Alok Maity
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yerbol Z Kurmangaliyev
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jesse Chin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Adam Z Stieg
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | | | - Sophia Hohlbauch
- Asylum Research, An Oxford Instruments Company, Santa Barbara, CA, 93117, USA
| | - Stefan Kaemmer
- Park Systems, 3040 Olcott St, Santa Clara, CA, 95054, USA
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - Thanh T Pham
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - Ken Yamauchi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Bennett G Novitch
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Roy Wollman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - April D Pyle
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
42
|
Claus C, Slavin M, Ansseau E, Lancelot C, Bah K, Lassche S, Fiévet M, Greco A, Tomaiuolo S, Tassin A, Dudome V, Kusters B, Declèves AE, Laoudj-Chenivesse D, van Engelen BGM, Nonclercq D, Belayew A, Kalisman N, Coppée F. The double homeodomain protein DUX4c is associated with regenerating muscle fibers and RNA-binding proteins. Skelet Muscle 2023; 13:5. [PMID: 36882853 PMCID: PMC9990282 DOI: 10.1186/s13395-022-00310-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/30/2022] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND We have previously demonstrated that double homeobox 4 centromeric (DUX4C) encoded for a functional DUX4c protein upregulated in dystrophic skeletal muscles. Based on gain- and loss-of-function studies we have proposed DUX4c involvement in muscle regeneration. Here, we provide further evidence for such a role in skeletal muscles from patients affected with facioscapulohumeral muscular dystrophy (FSHD). METHODS DUX4c was studied at RNA and protein levels in FSHD muscle cell cultures and biopsies. Its protein partners were co-purified and identified by mass spectrometry. Endogenous DUX4c was detected in FSHD muscle sections with either its partners or regeneration markers using co-immunofluorescence or in situ proximity ligation assay. RESULTS We identified new alternatively spliced DUX4C transcripts and confirmed DUX4c immunodetection in rare FSHD muscle cells in primary culture. DUX4c was detected in nuclei, cytoplasm or at cell-cell contacts between myocytes and interacted sporadically with specific RNA-binding proteins involved, a.o., in muscle differentiation, repair, and mass maintenance. In FSHD muscle sections, DUX4c was found in fibers with unusual shape or central/delocalized nuclei (a regeneration feature) staining for developmental myosin heavy chain, MYOD or presenting intense desmin labeling. Some couples of myocytes/fibers locally exhibited peripheral DUX4c-positive areas that were very close to each other, but in distinct cells. MYOD or intense desmin staining at these locations suggested an imminent muscle cell fusion. We further demonstrated DUX4c interaction with its major protein partner, C1qBP, inside myocytes/myofibers that presented features of regeneration. On adjacent muscle sections, we could unexpectedly detect DUX4 (the FSHD causal protein) and its interaction with C1qBP in fusing myocytes/fibers. CONCLUSIONS DUX4c upregulation in FSHD muscles suggests it contributes not only to the pathology but also, based on its protein partners and specific markers, to attempts at muscle regeneration. The presence of both DUX4 and DUX4c in regenerating FSHD muscle cells suggests DUX4 could compete with normal DUX4c functions, thus explaining why skeletal muscle is particularly sensitive to DUX4 toxicity. Caution should be exerted with therapeutic agents aiming for DUX4 suppression because they might also repress the highly similar DUX4c and interfere with its physiological role.
Collapse
Affiliation(s)
- Clothilde Claus
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Moriya Slavin
- Department of Biological Chemistry, the Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eugénie Ansseau
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Céline Lancelot
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Karimatou Bah
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Saskia Lassche
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands.,Department of Neurology, Zuyderland Medical Center, Heerlen, the Netherlands
| | - Manon Fiévet
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Anna Greco
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Sara Tomaiuolo
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium.,Laboratory of Respiratory Physiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Virginie Dudome
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Benno Kusters
- Department of Pathology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Anne-Emilie Declèves
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | | | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Denis Nonclercq
- Laboratory of Histology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Alexandra Belayew
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium
| | - Nir Kalisman
- Department of Biological Chemistry, the Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Frédérique Coppée
- Laboratory of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000, Mons, Belgium.
| |
Collapse
|
43
|
Hicks MR, Pyle AD. The emergence of the stem cell niche. Trends Cell Biol 2023; 33:112-123. [PMID: 35934562 PMCID: PMC9868094 DOI: 10.1016/j.tcb.2022.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 02/03/2023]
Abstract
Stem cell niches are composed of dynamic microenvironments that support stem cells over a lifetime. The emerging niche is distinct from the adult because its main role is to support the progenitors that build organ systems in development. Emerging niches mature through distinct stages to form the adult niche and enable proper stem cell support. As a model of emerging niches, this review highlights how differences in the skeletal muscle microenvironment influence emerging versus satellite cell (SC) niche formation in skeletal muscle, which is among the most regenerative tissue systems. We contrast how stem cell niches regulate intrinsic properties between progenitor and stem cells throughout development to adulthood. We describe new applications for generating emerging niches from human pluripotent stem cells (hPSCs) using developmental principles and highlight potential applications for regeneration and therapeutics.
Collapse
Affiliation(s)
- Michael R Hicks
- Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - April D Pyle
- Microbiology, Immunology, and Molecular Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
44
|
Togninalli M, Ho ATV, Madl CM, Holbrook CA, Wang YX, Magnusson KEG, Kirillova A, Chang A, Blau HM. Machine learning-based classification of dual fluorescence signals reveals muscle stem cell fate transitions in response to regenerative niche factors. NPJ Regen Med 2023; 8:4. [PMID: 36639373 PMCID: PMC9839750 DOI: 10.1038/s41536-023-00277-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
The proper regulation of muscle stem cell (MuSC) fate by cues from the niche is essential for regeneration of skeletal muscle. How pro-regenerative niche factors control the dynamics of MuSC fate decisions remains unknown due to limitations of population-level endpoint assays. To address this knowledge gap, we developed a dual fluorescence imaging time lapse (Dual-FLIT) microscopy approach that leverages machine learning classification strategies to track single cell fate decisions with high temporal resolution. Using two fluorescent reporters that read out maintenance of stemness and myogenic commitment, we constructed detailed lineage trees for individual MuSCs and their progeny, classifying each division event as symmetric self-renewing, asymmetric, or symmetric committed. Our analysis reveals that treatment with the lipid metabolite, prostaglandin E2 (PGE2), accelerates the rate of MuSC proliferation over time, while biasing division events toward symmetric self-renewal. In contrast, the IL6 family member, Oncostatin M (OSM), decreases the proliferation rate after the first generation, while blocking myogenic commitment. These insights into the dynamics of MuSC regulation by niche cues were uniquely enabled by our Dual-FLIT approach. We anticipate that similar binary live cell readouts derived from Dual-FLIT will markedly expand our understanding of how niche factors control tissue regeneration in real time.
Collapse
Affiliation(s)
- Matteo Togninalli
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Andrew T V Ho
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Department of Functional and Adaptive Biology - UMR 8251 CNRS, Université Paris Cité, 75013, Paris, France
| | - Christopher M Madl
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Colin A Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Yu Xin Wang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Klas E G Magnusson
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
- Department of Signal Processing, ACCESS Linnaeus Centre, KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Anna Kirillova
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Andrew Chang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, 94305-5175, USA.
| |
Collapse
|
45
|
Huerta M, Franco-Serrano L, Amela I, Perez-Pons JA, Piñol J, Mozo-Villarías A, Querol E, Cedano J. Role of Moonlighting Proteins in Disease: Analyzing the Contribution of Canonical and Moonlighting Functions in Disease Progression. Cells 2023; 12:cells12020235. [PMID: 36672169 PMCID: PMC9857295 DOI: 10.3390/cells12020235] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/09/2023] Open
Abstract
The term moonlighting proteins refers to those proteins that present alternative functions performed by a single polypeptide chain acquired throughout evolution (called canonical and moonlighting, respectively). Over 78% of moonlighting proteins are involved in human diseases, 48% are targeted by current drugs, and over 25% of them are involved in the virulence of pathogenic microorganisms. These facts encouraged us to study the link between the functions of moonlighting proteins and disease. We found a large number of moonlighting functions activated by pathological conditions that are highly involved in disease development and progression. The factors that activate some moonlighting functions take place only in pathological conditions, such as specific cellular translocations or changes in protein structure. Some moonlighting functions are involved in disease promotion while others are involved in curbing it. The disease-impairing moonlighting functions attempt to restore the homeostasis, or to reduce the damage linked to the imbalance caused by the disease. The disease-promoting moonlighting functions primarily involve the immune system, mesenchyme cross-talk, or excessive tissue proliferation. We often find moonlighting functions linked to the canonical function in a pathological context. Moonlighting functions are especially coordinated in inflammation and cancer. Wound healing and epithelial to mesenchymal transition are very representative. They involve multiple moonlighting proteins with a different role in each phase of the process, contributing to the current-phase phenotype or promoting a phase switch, mitigating the damage or intensifying the remodeling. All of this implies a new level of complexity in the study of pathology genesis, progression, and treatment. The specific protein function involved in a patient's progress or that is affected by a drug must be elucidated for the correct treatment of diseases.
Collapse
|
46
|
Jang H, Jo Y, Lee JH, Choi S. Aging of hair follicle stem cells and their niches. BMB Rep 2023; 56:2-9. [PMID: 36379515 PMCID: PMC9887102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/28/2023] Open
Abstract
Hair follicles in the skin undergo cyclic rounds of regeneration, degeneration, and rest throughout life. Stem cells residing in hair follicles play a pivotal role in maintaining tissue homeostasis and hair growth cycles. Research on hair follicle aging and age-related hair loss has demonstrated that a decline in hair follicle stem cell (HFSC) activity with aging can decrease the regeneration capacity of hair follicles. This review summarizes our understanding of how age-associated HFSC intrinsic and extrinsic mechanisms can induce HFSC aging and hair loss. In addition, we discuss approaches developed to attenuate ageassociated changes in HFSCs and their niches, thereby promoting hair regrowth. [BMB Reports 2023; 56(1): 2-9].
Collapse
Affiliation(s)
- Hansaem Jang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Yemin Jo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Jung Hyun Lee
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, Korea
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA, Incheon 21983, Korea
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
- School of Medical Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology (I_CREATE), Yonsei University, Incheon 21983, Korea
| |
Collapse
|
47
|
Jang H, Jo Y, Lee JH, Choi S. Aging of hair follicle stem cells and their niches. BMB Rep 2023; 56:2-9. [PMID: 36379515 PMCID: PMC9887102 DOI: 10.5483/bmbrep.2022-0183] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 09/06/2023] Open
Abstract
Hair follicles in the skin undergo cyclic rounds of regeneration, degeneration, and rest throughout life. Stem cells residing in hair follicles play a pivotal role in maintaining tissue homeostasis and hair growth cycles. Research on hair follicle aging and age-related hair loss has demonstrated that a decline in hair follicle stem cell (HFSC) activity with aging can decrease the regeneration capacity of hair follicles. This review summarizes our understanding of how age-associated HFSC intrinsic and extrinsic mechanisms can induce HFSC aging and hair loss. In addition, we discuss approaches developed to attenuate ageassociated changes in HFSCs and their niches, thereby promoting hair regrowth. [BMB Reports 2023; 56(1): 2-9].
Collapse
Affiliation(s)
- Hansaem Jang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Yemin Jo
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
| | - Jung Hyun Lee
- Division of Dermatology, Department of Medicine, University of Washington, Seattle, WA 98109, Korea
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA, Incheon 21983, Korea
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea, Incheon 21983, Korea
- School of Medical Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology (I_CREATE), Yonsei University, Incheon 21983, Korea
| |
Collapse
|
48
|
Chen W, Perkins TJ, Rudnicki MA. Quantification of Muscle Satellite Stem Cell Divisions by High-Content Analysis. Methods Mol Biol 2023; 2587:537-553. [PMID: 36401049 DOI: 10.1007/978-1-0716-2772-3_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
High-content screening is commonly performed on 2D cultured cells, which is high throughput but has low biological relevance. In contrast, single myofiber culture assay preserves the satellite cell niche between the basal lamina and sarcolemma and consequently has high biological relevance but is low throughput. We describe here a high-content screening method that utilizes single myofiber culture that addresses the caveats of both techniques. Our method utilizes the transgenic reporter allele Myf5-Cre:R26R-eYFP to differentiate stem and committed cells within a dividing couplet that can be quantified by high-content throughput immunodetection and bioinformatic analysis.
Collapse
Affiliation(s)
- William Chen
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Theodore J Perkins
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
49
|
Miyazaki A, Kawashima M, Nagata I, Miyoshi M, Miyakawa M, Sugiyama M, Sakuraya T, Sonomura T, Arakawa T. Icing after skeletal muscle injury decreases M1 macrophage accumulation and TNF-α expression during the early phase of muscle regeneration in rats. Histochem Cell Biol 2023; 159:77-89. [PMID: 36114866 DOI: 10.1007/s00418-022-02143-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
Following skeletal muscle injury, both myogenic and immune cells interact closely during the regenerative process. Although icing is still a common acute treatment for sports-related skeletal muscle injuries, icing after muscle injury has been shown to disrupt macrophage accumulation and impair muscle regeneration in animal models. However, it remains unknown whether icing shortly after injury affects macrophage-related phenomena during the early stages of muscle regeneration. Therefore, we focused on the distribution of M1/M2 macrophages and cytokines expressed predominantly by macrophages during the early stages of muscle regeneration after muscle crush injury. Icing resulted in a decrease, not retardation, in the accumulation of M1 macrophages, but not M2 macrophages, in injured muscles. Consistent with the decrease in M1 macrophage accumulation, icing led to a reduction, instead of delay, in the level of tumor necrosis factor-α (TNF-α) expression. Additionally, at subsequent timepoints, icing decreased the number of myogenic precursor cells in the regenerating area and the size of centrally nucleated regenerating myofibers. Together, our findings suggest that icing after acute muscle damage by crushing disturbs muscle regeneration through hindering tM1 macrophage-related phenomena.
Collapse
Affiliation(s)
- Anna Miyazaki
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Masato Kawashima
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.,Department of Health and Sports Science, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama, 701-0193, Japan
| | - Itsuki Nagata
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Makoto Miyoshi
- Department of Biophysics, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan
| | - Motoi Miyakawa
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.,Department of Health and Sport Sciences, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Megumi Sugiyama
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.,General Tokyo Hospital, 3-15-2 Egota, Nakano-ku, Tokyo, 165-8906, Japan
| | - Tohma Sakuraya
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.,Department of Oral Anatomy, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Takahiro Sonomura
- Department of Oral Anatomy, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Takamitsu Arakawa
- Department of Rehabilitation Sciences, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| |
Collapse
|
50
|
Montserrat-Vazquez S, Ali NJ, Matteini F, Lozano J, Zhaowei T, Mejia-Ramirez E, Marka G, Vollmer A, Soller K, Sacma M, Sakk V, Mularoni L, Mallm JP, Plass M, Zheng Y, Geiger H, Florian MC. Transplanting rejuvenated blood stem cells extends lifespan of aged immunocompromised mice. NPJ Regen Med 2022; 7:78. [PMID: 36581635 PMCID: PMC9800381 DOI: 10.1038/s41536-022-00275-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
One goal of regenerative medicine is to rejuvenate tissues and extend lifespan by restoring the function of endogenous aged stem cells. However, evidence that somatic stem cells can be targeted in vivo to extend lifespan is still lacking. Here, we demonstrate that after a short systemic treatment with a specific inhibitor of the small RhoGTPase Cdc42 (CASIN), transplanting aged hematopoietic stem cells (HSCs) from treated mice is sufficient to extend the healthspan and lifespan of aged immunocompromised mice without additional treatment. In detail, we show that systemic CASIN treatment improves strength and endurance of aged mice by increasing the myogenic regenerative potential of aged skeletal muscle stem cells. Further, we show that CASIN modifies niche localization and H4K16ac polarity of HSCs in vivo. Single-cell profiling reveals changes in HSC transcriptome, which underlie enhanced lymphoid and regenerative capacity in serial transplantation assays. Overall, we provide proof-of-concept evidence that a short systemic treatment to decrease Cdc42 activity improves the regenerative capacity of different endogenous aged stem cells in vivo, and that rejuvenated HSCs exert a broad systemic effect sufficient to extend murine health- and lifespan.
Collapse
Affiliation(s)
- Sara Montserrat-Vazquez
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Noelle J. Ali
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Francesca Matteini
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Lozano
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Tu Zhaowei
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Eva Mejia-Ramirez
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Gina Marka
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Angelika Vollmer
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Karin Soller
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Mehmet Sacma
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Vadim Sakk
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Loris Mularoni
- grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | | | - Mireya Plass
- grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain ,grid.417656.7Gene Regulation of Cell Identity Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Yi Zheng
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Hartmut Geiger
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - M. Carolina Florian
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|