101
|
García-Prat L, Perdiguero E, Alonso-Martín S, Dell'Orso S, Ravichandran S, Brooks SR, Juan AH, Campanario S, Jiang K, Hong X, Ortet L, Ruiz-Bonilla V, Flández M, Moiseeva V, Rebollo E, Jardí M, Sun HW, Musarò A, Sandri M, Del Sol A, Sartorelli V, Muñoz-Cánoves P. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age. Nat Cell Biol 2020; 22:1307-1318. [PMID: 33106654 DOI: 10.1038/s41556-020-00593-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.
Collapse
Affiliation(s)
- Laura García-Prat
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eusebio Perdiguero
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Sonia Alonso-Martín
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Neurosciences Area, Biodonostia Health Research Institute, Donostia-San Sebastián, San Sebastián, Spain
| | - Stefania Dell'Orso
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Srikanth Ravichandran
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Silvia Campanario
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Kan Jiang
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Xiaotong Hong
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Laura Ortet
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Vanessa Ruiz-Bonilla
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Marta Flández
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Grupo de Investigación en Oncología Clínico Traslacional, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Victoria Moiseeva
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Mercè Jardí
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Marco Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,CIC bioGUNE, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA.
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain. .,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
102
|
Khalil AS, Jaenisch R, Mooney DJ. Engineered tissues and strategies to overcome challenges in drug development. Adv Drug Deliv Rev 2020; 158:116-139. [PMID: 32987094 PMCID: PMC7518978 DOI: 10.1016/j.addr.2020.09.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/29/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022]
Abstract
Current preclinical studies in drug development utilize high-throughput in vitro screens to identify drug leads, followed by both in vitro and in vivo models to predict lead candidates' pharmacokinetic and pharmacodynamic properties. The goal of these studies is to reduce the number of lead drug candidates down to the most likely to succeed in later human clinical trials. However, only 1 in 10 drug candidates that emerge from preclinical studies will succeed and become an approved therapeutic. Lack of efficacy or undetected toxicity represents roughly 75% of the causes for these failures, despite these parameters being the primary exclusion criteria in preclinical studies. Recently, advances in both biology and engineering have created new tools for constructing new preclinical models. These models can complement those used in current preclinical studies by helping to create more realistic representations of human tissues in vitro and in vivo. In this review, we describe current preclinical models to identify their value and limitations and then discuss select areas of research where improvements in preclinical models are particularly needed to advance drug development. Following this, we discuss design considerations for constructing preclinical models and then highlight recent advances in these efforts. Taken together, we aim to review the advances as of 2020 surrounding the prospect of biological and engineering tools for adding enhanced biological relevance to preclinical studies to aid in the challenges of failed drug candidates and the burden this poses on the drug development enterprise and thus healthcare.
Collapse
Affiliation(s)
- Andrew S Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
103
|
Schutt C, Hallmann A, Hachim S, Klockner I, Valussi M, Atzberger A, Graumann J, Braun T, Boettger T. Linc-MYH configures INO80 to regulate muscle stem cell numbers and skeletal muscle hypertrophy. EMBO J 2020; 39:e105098. [PMID: 32960481 PMCID: PMC7667881 DOI: 10.15252/embj.2020105098] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/16/2022] Open
Abstract
Chromatin remodeling complexes have functions in transcriptional regulation and chromosome maintenance, but it is mostly unknown how the function of these normally ubiquitous complexes is specified in the cellular context. Here, we describe that the evolutionary conserved long non‐coding RNA linc‐MYH regulates the composition of the INO80 chromatin remodeler complex in muscle stem cells and prevents interaction with WDR5 and the transcription factor YY1. Linc‐MYH acts as a selective molecular switch in trans that governs the pro‐proliferative function of the ubiquitous INO80 complex but does not affect its role in maintaining genomic stability. The molecular switch is essential for restricting generation of quiescent MuSCs and proliferation of myoblasts in homeostasis and regeneration. Since linc‐MYH is expressed in proliferating myoblasts but not in quiescent MuSCs, we reason that the extent of myoblast proliferation has decisive effects on the size of the quiescent MuSC pool.
Collapse
Affiliation(s)
- Christian Schutt
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Alix Hallmann
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Salma Hachim
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Ina Klockner
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Melissa Valussi
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Ann Atzberger
- Max Planck Institute for Heart- and Lung Research, FACS Service Group, Bad Nauheim, Germany
| | - Johannes Graumann
- Max Planck Institute for Heart- and Lung Research, Mass Spectrometry Service Group, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| | - Thomas Boettger
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart- and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
104
|
Schüler SC, Gebert N, Ori A. Stem cell aging: The upcoming era of proteins and metabolites. Mech Ageing Dev 2020; 190:111288. [DOI: 10.1016/j.mad.2020.111288] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/04/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023]
|
105
|
Shwartz Y, Gonzalez-Celeiro M, Chen CL, Pasolli HA, Sheu SH, Fan SMY, Shamsi F, Assaad S, Lin ETY, Zhang B, Tsai PC, He M, Tseng YH, Lin SJ, Hsu YC. Cell Types Promoting Goosebumps Form a Niche to Regulate Hair Follicle Stem Cells. Cell 2020; 182:578-593.e19. [PMID: 32679029 DOI: 10.1016/j.cell.2020.06.031] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 04/06/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
Piloerection (goosebumps) requires concerted actions of the hair follicle, the arrector pili muscle (APM), and the sympathetic nerve, providing a model to study interactions across epithelium, mesenchyme, and nerves. Here, we show that APMs and sympathetic nerves form a dual-component niche to modulate hair follicle stem cell (HFSC) activity. Sympathetic nerves form synapse-like structures with HFSCs and regulate HFSCs through norepinephrine, whereas APMs maintain sympathetic innervation to HFSCs. Without norepinephrine signaling, HFSCs enter deep quiescence by down-regulating the cell cycle and metabolism while up-regulating quiescence regulators Foxp1 and Fgf18. During development, HFSC progeny secretes Sonic Hedgehog (SHH) to direct the formation of this APM-sympathetic nerve niche, which in turn controls hair follicle regeneration in adults. Our results reveal a reciprocal interdependence between a regenerative tissue and its niche at different stages and demonstrate sympathetic nerves can modulate stem cells through synapse-like connections and neurotransmitters to couple tissue production with demands.
Collapse
Affiliation(s)
- Yulia Shwartz
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Meryem Gonzalez-Celeiro
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Institute of Molecular Health Sciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Chih-Lung Chen
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - H Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Shu-Hsien Sheu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Sabrina Mai-Yi Fan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Steven Assaad
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Edrick Tai-Yu Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan
| | - Bing Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Pai-Chi Tsai
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Megan He
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Yu-Hua Tseng
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei 100, Taiwan; Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 100, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Ya-Chieh Hsu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
106
|
De Micheli AJ, Spector JA, Elemento O, Cosgrove BD. A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations. Skelet Muscle 2020; 10:19. [PMID: 32624006 PMCID: PMC7336639 DOI: 10.1186/s13395-020-00236-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) facilitates the unbiased reconstruction of multicellular tissue systems in health and disease. Here, we present a curated scRNA-seq dataset of human muscle samples from 10 adult donors with diverse anatomical locations. We integrated ~ 22,000 single-cell transcriptomes using Scanorama to account for technical and biological variation and resolved 16 distinct populations of muscle-resident cells using unsupervised clustering of the data compendium. These cell populations included muscle stem/progenitor cells (MuSCs), which bifurcated into discrete "quiescent" and "early-activated" MuSC subpopulations. Differential expression analysis identified transcriptional profiles altered in the activated MuSCs including genes associated with aging, obesity, diabetes, and impaired muscle regeneration, as well as long non-coding RNAs previously undescribed in human myogenic cells. Further, we modeled ligand-receptor cell-communication interactions and observed enrichment of the TWEAK-FN14 pathway in activated MuSCs, a characteristic signature of muscle wasting diseases. In contrast, the quiescent MuSCs have enhanced expression of the EGFR receptor, a recognized human MuSC marker. This work provides a new benchmark reference resource to examine human muscle tissue heterogeneity and identify potential targets in MuSC diversity and dysregulation in disease contexts.
Collapse
Affiliation(s)
- Andrea J De Micheli
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jason A Spector
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Division of Plastic Surgery, Weill Cornell Medical College, New York, NY, 10021, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
107
|
Henze H, Jung MJ, Ahrens HE, Steiner S, von Maltzahn J. Skeletal muscle aging – Stem cells in the spotlight. Mech Ageing Dev 2020; 189:111283. [DOI: 10.1016/j.mad.2020.111283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/04/2020] [Accepted: 06/01/2020] [Indexed: 01/09/2023]
|
108
|
Yue L, Wan R, Luan S, Zeng W, Cheung TH. Dek Modulates Global Intron Retention during Muscle Stem Cells Quiescence Exit. Dev Cell 2020; 53:661-676.e6. [DOI: 10.1016/j.devcel.2020.05.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/06/2020] [Accepted: 05/09/2020] [Indexed: 12/21/2022]
|
109
|
Abstract
Individuals that maintain healthy skeletal tissue tend to live healthier, happier lives as proper muscle function enables maintenance of independence and actuation of autonomy. The onset of skeletal muscle decline begins around the age of 30, and muscle atrophy is associated with a number of serious morbidities and mortalities. Satellite cells are responsible for regeneration of skeletal muscle and enter a reversible non-dividing state of quiescence under homeostatic conditions. In response to injury, satellite cells are able to activate and re-enter the cell cycle, creating new cells to repair and create nascent muscle fibres while preserving a small population that can return to quiescence for future regenerative demands. However, in aged muscle, satellite cells that experience prolonged quiescence will undergo programmed cellular senescence, an irreversible non-dividing state that handicaps the regenerative capabilities of muscle. This review examines how periodic activation and cycling of satellite cells through exercise can mitigate senescence acquisition and myogenic decline.
Collapse
Affiliation(s)
- William Chen
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - David Datzkiw
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada K1H 8L6.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada K1H 8M5
| |
Collapse
|
110
|
Xi H, Langerman J, Sabri S, Chien P, Young CS, Younesi S, Hicks M, Gonzalez K, Fujiwara W, Marzi J, Liebscher S, Spencer M, Van Handel B, Evseenko D, Schenke-Layland K, Plath K, Pyle AD. A Human Skeletal Muscle Atlas Identifies the Trajectories of Stem and Progenitor Cells across Development and from Human Pluripotent Stem Cells. Cell Stem Cell 2020; 27:158-176.e10. [PMID: 32396864 PMCID: PMC7367475 DOI: 10.1016/j.stem.2020.04.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 03/12/2020] [Accepted: 04/23/2020] [Indexed: 12/17/2022]
Abstract
The developmental trajectory of human skeletal myogenesis and the transition between progenitor and stem cell states are unclear. We used single-cell RNA sequencing to profile human skeletal muscle tissues from embryonic, fetal, and postnatal stages. In silico, we identified myogenic as well as other cell types and constructed a "roadmap" of human skeletal muscle ontogeny across development. In a similar fashion, we also profiled the heterogeneous cell cultures generated from multiple human pluripotent stem cell (hPSC) myogenic differentiation protocols and mapped hPSC-derived myogenic progenitors to an embryonic-to-fetal transition period. We found differentially enriched biological processes and discovered co-regulated gene networks and transcription factors present at distinct myogenic stages. This work serves as a resource for advancing our knowledge of human myogenesis. It also provides a tool for a better understanding of hPSC-derived myogenic progenitors for translational applications in skeletal muscle-based regenerative medicine.
Collapse
Affiliation(s)
- Haibin Xi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Justin Langerman
- Deparment of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shan Sabri
- Deparment of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peggie Chien
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Courtney S Young
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shahab Younesi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Hicks
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Karen Gonzalez
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wakana Fujiwara
- Department of Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Julia Marzi
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany; The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Simone Liebscher
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Melissa Spencer
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ben Van Handel
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, Stem Cell Research and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, Tübingen, Germany; The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany; Department of Medicine/Cardiology, Cardiovascular Research Laboratories, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kathrin Plath
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Deparment 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; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| | - April D Pyle
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
111
|
Kimmel JC, Hwang AB, Scaramozza A, Marshall WF, Brack AS. Aging induces aberrant state transition kinetics in murine muscle stem cells. Development 2020; 147:dev183855. [PMID: 32198156 PMCID: PMC7225128 DOI: 10.1242/dev.183855] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022]
Abstract
Murine muscle stem cells (MuSCs) experience a transition from quiescence to activation that is required for regeneration, but it remains unknown if the trajectory and dynamics of activation change with age. Here, we use time-lapse imaging and single cell RNA-seq to measure activation trajectories and rates in young and aged MuSCs. We find that the activation trajectory is conserved in aged cells, and we develop effective machine-learning classifiers for cell age. Using cell-behavior analysis and RNA velocity, we find that activation kinetics are delayed in aged MuSCs, suggesting that changes in stem cell dynamics may contribute to impaired stem cell function with age. Intriguingly, we also find that stem cell activation appears to be a random walk-like process, with frequent reversals, rather than a continuous linear progression. These results support a view of the aged stem cell phenotype as a combination of differences in the location of stable cell states and differences in transition rates between them.
Collapse
Affiliation(s)
- Jacob C Kimmel
- Eli and Edythe Broad Center for Regenerative Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, CA 94143, USA
- Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ara B Hwang
- Eli and Edythe Broad Center for Regenerative Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
| | - Annarita Scaramozza
- Eli and Edythe Broad Center for Regenerative Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
| | - Wallace F Marshall
- Center for Cellular Construction, University of California, San Francisco, San Francisco, CA 94143, USA
- Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrew S Brack
- Eli and Edythe Broad Center for Regenerative Medicine, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
| |
Collapse
|
112
|
Gebert N, Cheng CW, Kirkpatrick JM, Di Fraia D, Yun J, Schädel P, Pace S, Garside GB, Werz O, Rudolph KL, Jasper H, Yilmaz ÖH, Ori A. Region-Specific Proteome Changes of the Intestinal Epithelium during Aging and Dietary Restriction. Cell Rep 2020; 31:107565. [PMID: 32348758 PMCID: PMC7446723 DOI: 10.1016/j.celrep.2020.107565] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/05/2020] [Accepted: 04/02/2020] [Indexed: 01/18/2023] Open
Abstract
The small intestine is responsible for nutrient absorption and one of the most important interfaces between the environment and the body. During aging, changes of the epithelium lead to food malabsorption and reduced barrier function, thus increasing disease risk. The drivers of these alterations remain poorly understood. Here, we compare the proteomes of intestinal crypts from mice across different anatomical regions and ages. We find that aging alters epithelial immunity, metabolism, and cell proliferation and is accompanied by region-dependent skewing in the cellular composition of the epithelium. Of note, short-term dietary restriction followed by refeeding partially restores the epithelium by promoting stem cell differentiation toward the secretory lineage. We identify Hmgcs2 (3-hydroxy-3-methylglutaryl-coenzyme A [CoA] synthetase 2), the rate-limiting enzyme for ketogenesis, as a modulator of stem cell differentiation that responds to dietary changes, and we provide an atlas of region- and age-dependent proteome changes of the small intestine.
Collapse
Affiliation(s)
- Nadja Gebert
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Chia-Wei Cheng
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | | | - Domenico Di Fraia
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Jina Yun
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Patrick Schädel
- Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - Simona Pace
- Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - George B Garside
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Oliver Werz
- Institute of Pharmacy, Friedrich Schiller University, Jena, Germany
| | - K Lenhard Rudolph
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Henri Jasper
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ömer H Yilmaz
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
| |
Collapse
|
113
|
Oprescu SN, Yue F, Qiu J, Brito LF, Kuang S. Temporal Dynamics and Heterogeneity of Cell Populations during Skeletal Muscle Regeneration. iScience 2020; 23:100993. [PMID: 32248062 PMCID: PMC7125354 DOI: 10.1016/j.isci.2020.100993] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/08/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Mammalian skeletal muscle possesses a unique ability to regenerate, which is primarily mediated by a population of resident muscle stem cells (MuSCs) and requires a concerted response from other supporting cell populations. Previous targeted analysis has described the involvement of various specific populations in regeneration, but an unbiased and simultaneous evaluation of all cell populations has been limited. Therefore, we used single-cell RNA-sequencing to uncover gene expression signatures of over 53,000 individual cells during skeletal muscle regeneration. Cells clustered into 25 populations and subpopulations, including a subpopulation of immune gene enriched myoblasts (immunomyoblasts) and subpopulations of fibro-adipogenic progenitors. Our analyses also uncovered striking spatiotemporal dynamics in gene expression, population composition, and cell-cell interaction during muscle regeneration. These findings provide insights into the cellular and molecular underpinning of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, 915 W State St, West Lafayette, IN 47907, USA
| | - Feng Yue
- Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Biological Sciences, Purdue University, 915 W State St, West Lafayette, IN 47907, USA; Department of Animal Sciences, Purdue University, 270 S Russell St, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, 201 S University St, West Lafayette, IN 47907, USA.
| |
Collapse
|
114
|
Englund DA, Murach KA, Dungan CM, Figueiredo VC, Vechetti IJ, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Depletion of resident muscle stem cells negatively impacts running volume, physical function, and muscle fiber hypertrophy in response to lifelong physical activity. Am J Physiol Cell Physiol 2020; 318:C1178-C1188. [PMID: 32320286 DOI: 10.1152/ajpcell.00090.2020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To date, studies that have aimed to investigate the role of satellite cells during adult skeletal muscle adaptation and hypertrophy have utilized a nontranslational stimulus and/or have been performed over a relatively short time frame. Although it has been shown that satellite cell depletion throughout adulthood does not drive skeletal muscle loss in sedentary mice, it remains unknown how satellite cells participate in skeletal muscle adaptation to long-term physical activity. The current study was designed to determine whether reduced satellite cell content throughout adulthood would influence the transcriptome-wide response to physical activity and diminish the adaptive response of skeletal muscle. We administered vehicle or tamoxifen to adult Pax7-diphtheria toxin A (DTA) mice to deplete satellite cells and assigned them to sedentary or wheel-running conditions for 13 mo. Satellite cell depletion throughout adulthood reduced balance and coordination, overall running volume, and the size of muscle proprioceptors (spindle fibers). Furthermore, satellite cell participation was necessary for optimal muscle fiber hypertrophy but not adaptations in fiber type distribution in response to lifelong physical activity. Transcriptome-wide analysis of the plantaris and soleus revealed that satellite cell function is muscle type specific; satellite cell-dependent myonuclear accretion was apparent in oxidative muscles, whereas initiation of G protein-coupled receptor (GPCR) signaling in the glycolytic plantaris may require satellite cells to induce optimal adaptations to long-term physical activity. These findings suggest that satellite cells play a role in preserving physical function during aging and influence muscle adaptation during sustained periods of physical activity.
Collapse
Affiliation(s)
- Davis A Englund
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Kevin A Murach
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Cory M Dungan
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Vandré C Figueiredo
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - Ivan J Vechetti
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Esther E Dupont-Versteegden
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| | - John J McCarthy
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Charlotte A Peterson
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky.,Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
115
|
Dungan CM, Peck BD, Walton RG, Huang Z, Bamman MM, Kern PA, Peterson CA. In vivo analysis of γH2AX+ cells in skeletal muscle from aged and obese humans. FASEB J 2020; 34:7018-7035. [PMID: 32246795 DOI: 10.1096/fj.202000111rr] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/25/2022]
Abstract
Over the past 20 years, various identifiers of cellular senescence have been used to quantify the abundance of these cells in different tissues. These include classic markers such as p16, senescence-associated β-gal, and γH2AX, in addition to more recent markers (Sudan Black B and HMGB1). In vivo data on the usefulness of these markers in skeletal muscle are very limited and inconsistent. In the present study, we attempted to identify senescent cells in frozen human skeletal muscle biopsies using these markers to determine the effects of age and obesity on senescent cell burden; however, we were only able to assess the abundance of DNA-damaged nuclei using γH2AX immunohistochemistry. The abundance of γH2AX+ cells, including satellite cells, was not higher in muscle from old compared to young individuals; however, γH2AX+ cells were higher with obesity. Additionally, terminally differentiated, postmitotic myofiber nuclei from obese individuals had elevated γH2AX abundance compared to muscle from lean individuals. Analyses of gene expression support the conclusion that the elevated DNA damage and the senescence-associated secretory phenotype are preferentially associated with obesity in skeletal muscle. These data implicate obesity as a larger contributor to DNA damage in skeletal muscle than aging; however, more sensitive senescence markers for human skeletal muscle are needed to determine if these cells are in fact senescent.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Bailey D Peck
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - R Grace Walton
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| | - Zhengyan Huang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Marcas M Bamman
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama, Birmingham, AL, USA.,Geriatric Research, Education, and Clinical Center, Birmingham VA Medical Center, Birmingham, AL, USA
| | - Philip A Kern
- Department of Internal Medicine/Endocrinology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Charlotte A Peterson
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, KY, USA.,Center for Muscle Biology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
116
|
Barruet E, Garcia SM, Striedinger K, Wu J, Lee S, Byrnes L, Wong A, Xuefeng S, Tamaki S, Brack AS, Pomerantz JH. Functionally heterogeneous human satellite cells identified by single cell RNA sequencing. eLife 2020; 9:51576. [PMID: 32234209 PMCID: PMC7164960 DOI: 10.7554/elife.51576] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
Although heterogeneity is recognized within the murine satellite cell pool, a comprehensive understanding of distinct subpopulations and their functional relevance in human satellite cells is lacking. We used a combination of single cell RNA sequencing and flow cytometry to identify, distinguish, and physically separate novel subpopulations of human PAX7+ satellite cells (Hu-MuSCs) from normal muscles. We found that, although relatively homogeneous compared to activated satellite cells and committed progenitors, the Hu-MuSC pool contains clusters of transcriptionally distinct cells with consistency across human individuals. New surface marker combinations were enriched in transcriptional subclusters, including a subpopulation of Hu-MuSCs marked by CXCR4/CD29/CD56/CAV1 (CAV1+). In vitro, CAV1+ Hu-MuSCs are morphologically distinct, and characterized by resistance to activation compared to CAV1- Hu-MuSCs. In vivo, CAV1+ Hu-MuSCs demonstrated increased engraftment after transplantation. Our findings provide a comprehensive transcriptional view of normal Hu-MuSCs and describe new heterogeneity, enabling separation of functionally distinct human satellite cell subpopulations.
Collapse
Affiliation(s)
- Emilie Barruet
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Steven M Garcia
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Katharine Striedinger
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Jake Wu
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Solomon Lee
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Lauren Byrnes
- University of California San Francisco, San Francisco, United States
| | - Alvin Wong
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Sun Xuefeng
- Department of Orthopedic Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Stanley Tamaki
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Andrew S Brack
- Department of Orthopedic Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| | - Jason H Pomerantz
- Departments of Surgery and Orofacial Sciences, Division of Plastic and Reconstructive Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
117
|
Strenzke M, Alberton P, Aszodi A, Docheva D, Haas E, Kammerlander C, Böcker W, Saller MM. Tenogenic Contribution to Skeletal Muscle Regeneration: The Secretome of Scleraxis Overexpressing Mesenchymal Stem Cells Enhances Myogenic Differentiation In Vitro. Int J Mol Sci 2020; 21:E1965. [PMID: 32183051 PMCID: PMC7139530 DOI: 10.3390/ijms21061965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 01/07/2023] Open
Abstract
Integrity of the musculoskeletal system is essential for the transfer of muscular contraction force to the associated bones. Tendons and skeletal muscles intertwine, but on a cellular level, the myotendinous junctions (MTJs) display a sharp transition zone with a highly specific molecular adaption. The function of MTJs could go beyond a mere structural role and might include homeostasis of this musculoskeletal tissue compound, thus also being involved in skeletal muscle regeneration. Repair processes recapitulate several developmental mechanisms, and as myotendinous interaction does occur already during development, MTJs could likewise contribute to muscle regeneration. Recent studies identified tendon-related, scleraxis-expressing cells that reside in close proximity to the MTJs and the muscle belly. As the muscle-specific function of these scleraxis positive cells is unknown, we compared the influence of two immortalized mesenchymal stem cell (MSC) lines-differing only by the overexpression of scleraxis-on myoblasts morphology, metabolism, migration, fusion, and alignment. Our results revealed a significant increase in myoblast fusion and metabolic activity when exposed to the secretome derived from scleraxis-overexpressing MSCs. However, we found no significant changes in myoblast migration and myofiber alignment. Further analysis of differentially expressed genes between native MSCs and scleraxis-overexpressing MSCs by RNA sequencing unraveled potential candidate genes, i.e., extracellular matrix (ECM) proteins, transmembrane receptors, or proteases that might enhance myoblast fusion. Our results suggest that musculotendinous interaction is essential for the development and healing of skeletal muscles.
Collapse
Affiliation(s)
- Maximilian Strenzke
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany;
| | - Elisabeth Haas
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Christian Kammerlander
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Wolfgang Böcker
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| |
Collapse
|
118
|
An optimized chemical-genetic method for cell-specific metabolic labeling of RNA. Nat Methods 2020; 17:311-318. [PMID: 32015544 PMCID: PMC8518020 DOI: 10.1038/s41592-019-0726-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
Tissues and organs are composed of diverse cell types, which poses a major challenge for cell-specific gene expression profiling. Current metabolic labeling methods rely on the inability of mammalian cells to incorporate exogenous pyrimidine analogs, which are then co-opted by ectopically-expressed enzymes. We demonstrate that mammalian cells can incorporate uracil analogs and characterize the enzymatic pathways responsible for high background incorporation. To overcome these limitations, we developed a novel small-molecule/enzyme pair consisting of uridine-cytidine kinase 2 (UCK2) and 2’-azidouridine (2’AzUd). We demonstrate that 2’AzUd is only incorporated in UCK2-expressing cells and characterize selectivity mechanisms using molecular dynamics and X-ray crystallography. Furthermore, this pair can be used to purify and track RNA from specific cellular populations, making it ideal for high-resolution cell-specific RNA labeling. Overall, these results reveal novel aspects of mammalian salvage pathways and serve as a new benchmark for designing, characterizing and evaluating cell-specific biomolecule labeling methodologies.
Collapse
|
119
|
Choi IY, Lim H, Cho HJ, Oh Y, Chou BK, Bai H, Cheng L, Kim YJ, Hyun S, Kim H, Shin JH, Lee G. Transcriptional landscape of myogenesis from human pluripotent stem cells reveals a key role of TWIST1 in maintenance of skeletal muscle progenitors. eLife 2020; 9:e46981. [PMID: 32011235 PMCID: PMC6996923 DOI: 10.7554/elife.46981] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
Generation of skeletal muscle cells with human pluripotent stem cells (hPSCs) opens new avenues for deciphering essential, but poorly understood aspects of transcriptional regulation in human myogenic specification. In this study, we characterized the transcriptional landscape of distinct human myogenic stages, including OCT4::EGFP+ pluripotent stem cells, MSGN1::EGFP+ presomite cells, PAX7::EGFP+ skeletal muscle progenitor cells, MYOG::EGFP+ myoblasts, and multinucleated myotubes. We defined signature gene expression profiles from each isolated cell population with unbiased clustering analysis, which provided unique insights into the transcriptional dynamics of human myogenesis from undifferentiated hPSCs to fully differentiated myotubes. Using a knock-out strategy, we identified TWIST1 as a critical factor in maintenance of human PAX7::EGFP+ putative skeletal muscle progenitor cells. Our data revealed a new role of TWIST1 in human skeletal muscle progenitors, and we have established a foundation to identify transcriptional regulations of human myogenic ontogeny (online database can be accessed in http://www.myogenesis.net/).
Collapse
Affiliation(s)
- In Young Choi
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Department of Medicine, Graduate SchoolKyung Hee UniversitySeoulRepublic of Korea
| | - Hotae Lim
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- College of Veterinary MedicineChungbuk National UniversityChungbukRepublic of Korea
| | - Hyeon Jin Cho
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Yohan Oh
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Bin-Kuan Chou
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Division of Hematology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Hao Bai
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Division of Hematology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Linzhao Cheng
- Division of Hematology, Department of MedicineJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Yong Jun Kim
- Department of Pathololgy, College of MedicineKyung Hee UniversitySeoulRepublic of Korea
| | - SangHwan Hyun
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- College of Veterinary MedicineChungbuk National UniversityChungbukRepublic of Korea
| | - Hyesoo Kim
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Department of NeurologyJohns Hopkins University, School of MedicineBaltimoreUnited States
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical CampusBaltimoreUnited States
| | - Gabsang Lee
- The Institute for Cell EngineeringJohns Hopkins University, School of MedicineBaltimoreUnited States
- Department of NeurologyJohns Hopkins University, School of MedicineBaltimoreUnited States
- The Solomon H. Synder Department of NeuroscienceJohns Hopkins University, School of MedicineBaltimoreUnited States
| |
Collapse
|
120
|
Cable J, Fuchs E, Weissman I, Jasper H, Glass D, Rando TA, Blau H, Debnath S, Oliva A, Park S, Passegué E, Kim C, Krasnow MA. Adult stem cells and regenerative medicine-a symposium report. Ann N Y Acad Sci 2020; 1462:27-36. [PMID: 31655007 PMCID: PMC7135961 DOI: 10.1111/nyas.14243] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
Adult stem cells are rare, undifferentiated cells found in all tissues of the body. Although normally kept in a quiescent, nondividing state, these cells can proliferate and differentiate to replace naturally dying cells within their tissue and to repair its wounds in response to injury. Due to their proliferative nature and ability to regenerate tissue, adult stem cells have the potential to treat a variety of degenerative diseases as well as aging. In addition, since stem cells are often thought to be the source of malignant tumors, understanding the mechanisms that keep their proliferative abilities in check can pave the way for new cancer therapies. While adult stem cells have had limited practical and clinical applications to date, several clinical trials of stem cell-based therapies are underway. This report details recent research presented at the New York Academy of Sciences on March 14, 2019 on understanding the factors that regulate stem cell activity and differentiation, with the hope of translating these findings into the clinic.
Collapse
Affiliation(s)
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, New York
| | - Irving Weissman
- Pathology Stem Cell Institute, Stanford University, Stanford, California
| | | | | | - Thomas A Rando
- Neurology & Neurological Sciences, Stanford University, Stanford, California
| | - Helen Blau
- Microbiology and Immunology - Baxter Labs, Stanford University, Stanford, California
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | | | - Sangbum Park
- Department of Genetics, Yale University, New Haven, Connecticut
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics & Development, Columbia University, New York, New York
| | - Carla Kim
- Dana Farber/Harvard Cancer Center, Boston, Massachusetts
- Division of Hematology/Oncology, Stem Cell Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts
- Genetics Department, Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Mark A Krasnow
- Department of Biochemistry, Stanford University, Stanford, California
| |
Collapse
|
121
|
Feige P, Rudnicki MA. Isolation of satellite cells and transplantation into mice for lineage tracing in muscle. Nat Protoc 2020; 15:1082-1097. [PMID: 31965111 DOI: 10.1038/s41596-019-0278-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/03/2019] [Indexed: 02/03/2023]
Abstract
Limited methods exist to assay the direct effects of therapeutic intervention on muscle stem cell fate, proliferation or differentiation in an in vivo context. Here we provide an optimized protocol for muscle stem cell isolation and transplantation into mice to deconvolute heterogeneity within isolated stem cell populations. Viable and pure cell populations are isolated within 2 h and can then be used for therapeutic intervention or transplantation to uncover the repopulating and differentiation potential in mice, a physiologically relevant in vivo context. Effects can be assessed 9 d after transplantation. This methodology analyzes cell and sort purity prior to transplantation to improve reproducibility and outlines novel blocking steps to improve tissue staining and analysis. Experience with surgical procedures in mice is recommended before attempting this protocol. Our system is widely applicable for exploring stem cell dynamics within muscle and has already been used to study heterogeneity within muscle stem cell populations and efficacy of therapeutic intervention on isolated stem cell populations.
Collapse
Affiliation(s)
- Peter Feige
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, 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
|
122
|
Abstract
Satellite cells are the main muscle-resident cells responsible for muscle regeneration. Much research has described this population as being heterogeneous, but little is known about the different roles each subpopulation plays. Recent advances in the field have utilized the power of single-cell analysis to better describe and functionally characterize subpopulations of satellite cells as well as other cell groups comprising the muscle tissue. Furthermore, emerging technologies are opening the door to answering as-yet-unresolved questions pertaining to satellite cell heterogeneity and cell fate decisions.
Collapse
Affiliation(s)
- John Saber
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Alexander Y.T. Lin
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Michael A. Rudnicki
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| |
Collapse
|
123
|
Boscolo Sesillo F, Wong M, Cortez A, Alperin M. Isolation of muscle stem cells from rat skeletal muscles. Stem Cell Res 2019; 43:101684. [PMID: 31931473 PMCID: PMC7357689 DOI: 10.1016/j.scr.2019.101684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/15/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023] Open
Abstract
Muscle stem cells (MuSCs) are involved in homeostatic maintenance of skeletal muscle and play a central role in muscle regeneration in response to injury. Thus, understanding MuSC autonomous properties is of fundamental importance for studies of muscle degenerative diseases and muscle plasticity. Rat, as an animal model, has been widely used in the skeletal muscle field, however rat MuSC isolation through fluorescence-activated cell sorting has never been described. This work validates a protocol for effective MuSC isolation from rat skeletal muscles. Tibialis anterior was harvested from female rats and digested for isolation of MuSCs. Three protocols, employing different cell surface markers (CD106, CD56, and CD29), were compared for their ability to isolate a highly enriched MuSC population. Cells isolated using only CD106 as a positive marker showed high expression of Pax7, ability to progress through myogenic lineage while in culture, and complete differentiation in serum-deprived conditions. The protocol was further validated in gastrocnemius, diaphragm, and the individual components of the pelvic floor muscle complex (coccygeus, iliocaudalis, and pubocaudalis), proving to be reproducible. CD106 is an efficient marker for reliable isolation of MuSCs from a variety of rat skeletal muscles.
Collapse
Affiliation(s)
- Francesca Boscolo Sesillo
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Michelle Wong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California San Diego, La Jolla, CA 92093, USA
| | - Amy Cortez
- Flow Cytometry Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marianna Alperin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
124
|
Kann AP, Krauss RS. Multiplexed RNAscope and immunofluorescence on whole-mount skeletal myofibers and their associated stem cells. Development 2019; 146:dev179259. [PMID: 31519691 PMCID: PMC6826044 DOI: 10.1242/dev.179259] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022]
Abstract
Skeletal muscle myofibers are large syncytial cells comprising hundreds of myonuclei, and in situ hybridization experiments have reported a range of transcript localization patterns within them. Although some transcripts are uniformly distributed throughout myofibers, proximity to specialized regions can affect the programming of myonuclei and functional compartmentalization of transcripts. Established techniques are limited by a lack of both sensitivity and spatial resolution, restricting the ability to identify different patterns of gene expression. In this study, we adapted RNAscope fluorescent in situ hybridization technology for use on whole-mount mouse primary myofibers, a preparation that isolates single myofibers with their associated muscle stem cells remaining in their niche. This method can be combined with immunofluorescence, enabling an unparalleled ability to visualize and quantify transcripts and proteins across the length and depth of skeletal myofibers and their associated stem cells. Using this approach, we demonstrate a range of potential uses, including the visualization of specialized transcriptional programming within myofibers, tracking activation-induced transcriptional changes, quantification of stem cell heterogeneity and evaluation of stem cell niche factor transcription patterns.
Collapse
Affiliation(s)
- Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
125
|
Gene expression profiling of skeletal myogenesis in human embryonic stem cells reveals a potential cascade of transcription factors regulating stages of myogenesis, including quiescent/activated satellite cell-like gene expression. PLoS One 2019; 14:e0222946. [PMID: 31560727 PMCID: PMC6764674 DOI: 10.1371/journal.pone.0222946] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/10/2019] [Indexed: 01/05/2023] Open
Abstract
Human embryonic stem cell (hESC)-derived skeletal muscle progenitors (SMP)—defined as PAX7-expressing cells with myogenic potential—can provide an abundant source of donor material for muscle stem cell therapy. As in vitro myogenesis is decoupled from in vivo timing and 3D-embryo structure, it is important to characterize what stage or type of muscle is modeled in culture. Here, gene expression profiling is analyzed in hESCs over a 50 day skeletal myogenesis protocol and compared to datasets of other hESC-derived skeletal muscle and adult murine satellite cells. Furthermore, day 2 cultures differentiated with high or lower concentrations of CHIR99021, a GSK3A/GSK3B inhibitor, were contrasted. Expression profiling of the 50 day time course identified successively expressed gene subsets involved in mesoderm/paraxial mesoderm induction, somitogenesis, and skeletal muscle commitment/formation which could be regulated by a putative cascade of transcription factors. Initiating differentiation with higher CHIR99021 concentrations significantly increased expression of MSGN1 and TGFB-superfamily genes, notably NODAL, resulting in enhanced paraxial mesoderm and reduced ectoderm/neuronal gene expression. Comparison to adult satellite cells revealed that genes expressed in 50-day cultures correlated better with those expressed by quiescent or early activated satellite cells, which have the greatest therapeutic potential. Day 50 cultures were similar to other hESC-derived skeletal muscle and both expressed known and novel SMP surface proteins. Overall, a putative cascade of transcription factors has been identified which regulates four stages of myogenesis. Subsets of these factors were upregulated by high CHIR99021 or their binding sites were significantly over-represented during SMP activation, ranging from quiescent to late-activated stages. This analysis serves as a resource to further study the progression of in vitro skeletal myogenesis and could be mined to identify novel markers of pluripotent-derived SMPs or regulatory transcription/growth factors. Finally, 50-day hESC-derived SMPs appear similar to quiescent/early activated satellite cells, suggesting they possess therapeutic potential.
Collapse
|
126
|
Wnt4 from the Niche Controls the Mechano-Properties and Quiescent State of Muscle Stem Cells. Cell Stem Cell 2019; 25:654-665.e4. [PMID: 31495781 DOI: 10.1016/j.stem.2019.08.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 04/19/2019] [Accepted: 08/12/2019] [Indexed: 12/31/2022]
Abstract
Satellite cells (SCs) reside in a dormant state during tissue homeostasis. The specific paracrine agents and niche cells that maintain SC quiescence remain unknown. We find that Wnt4 produced by the muscle fiber maintains SC quiescence through RhoA. Using cell-specific inducible genetics, we find that a Wnt4-Rho signaling axis constrains SC numbers and activation during tissue homeostasis in adult mice. Wnt4 activates Rho in quiescent SCs to maintain mechanical strain, restrict movement in the niche, and repress YAP. The induction of YAP upon disruption of RhoA is essential for SC activation under homeostasis. In the context of injury, the loss of Wnt4 from the niche accelerates SC activation and muscle repair, whereas overexpression of Wnt4 transitions SCs into a deeper state of quiescence and delays muscle repair. In conclusion, the SC pool undergoes dynamic transitions during early activation with changes in mechano-properties and cytoskeleton signaling preceding cell-cycle entry.
Collapse
|
127
|
Potter SS. Single-cell RNA sequencing for the study of development, physiology and disease. Nat Rev Nephrol 2019; 14:479-492. [PMID: 29789704 DOI: 10.1038/s41581-018-0021-7] [Citation(s) in RCA: 377] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An ongoing technological revolution is continually improving our ability to carry out very high-resolution studies of gene expression patterns. Current technology enables the global gene expression profiles of single cells to be defined, facilitating dissection of heterogeneity in cell populations that was previously hidden. In contrast to gene expression studies that use bulk RNA samples and provide only a virtual average of the diverse constituent cells, single-cell studies enable the molecular distinction of all cell types within a complex population mix, such as a tumour or developing organ. For instance, single-cell gene expression profiling has contributed to improved understanding of how histologically identical, adjacent cells make different differentiation decisions during development. Beyond development, single-cell gene expression studies have enabled the characteristics of previously known cell types to be more fully defined and facilitated the identification of novel categories of cells, contributing to improvements in our understanding of both normal and disease-related physiological processes and leading to the identification of new treatment approaches. Although limitations remain to be overcome, technology for the analysis of single-cell gene expression patterns is improving rapidly and beginning to provide a detailed atlas of the gene expression patterns of all cell types in the human body.
Collapse
Affiliation(s)
- S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
128
|
Greenblatt MB, Ono N, Ayturk UM, Debnath S, Lalani S. The Unmixing Problem: A Guide to Applying Single-Cell RNA Sequencing to Bone. J Bone Miner Res 2019; 34:1207-1219. [PMID: 31336008 PMCID: PMC6658136 DOI: 10.1002/jbmr.3802] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 12/24/2022]
Abstract
Bone is composed of a complex mixture of many dynamic cell types. Flow cytometry and in vivo lineage tracing have offered early progress toward deconvoluting this heterogeneous mixture of cells into functionally well-defined populations suitable for further studies. Single-cell sequencing is poised as a key complementary technique to better understand the cellular basis of bone metabolism and development. However, single-cell sequencing approaches still have important limitations, including transcriptional effects of cell isolation and sparse sampling of the transcriptome, that must be considered during experimental design and analysis to harness the power of this approach. Accounting for these limitations requires a deep knowledge of the tissue under study. Therefore, with the emergence of accessible tools for conducting and analyzing single-cell RNA sequencing (scRNA-seq) experiments, bone biologists will be ideal leaders in the application of scRNA-seq to the skeleton. Here we provide an overview of the steps involved with a single-cell sequencing analysis of bone, focusing on practical considerations needed for a successful study. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, USA
- Research Division, Hospital for Special Surgery, New York,
NY, USA
| | - Noriaki Ono
- University of Michigan School of Dentistry, Ann Arbor, MI,
USA
| | - Ugur M Ayturk
- Musculoskeletal Integrity Program, Hospital for Special
Surgery, New York, NY, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, USA
| | - Sarfaraz Lalani
- Department of Pathology and Laboratory Medicine, Weill
Cornell Medicine, New York, NY, USA
| |
Collapse
|
129
|
Maguire G, Paler L, Green L, Mella R, Valcarcel M, Villace P. Rescue of degenerating neurons and cells by stem cell released molecules: using a physiological renormalization strategy. Physiol Rep 2019; 7:e14072. [PMID: 31050222 PMCID: PMC6497969 DOI: 10.14814/phy2.14072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/26/2019] [Accepted: 03/31/2019] [Indexed: 12/13/2022] Open
Abstract
Evidence suggests that adult stem cell types and progenitor cells act collectively in a given tissue to maintain and heal organs, such as muscle, through a release of a multitude of molecules packaged into exosomes from the different cell types. Using this principle for the development of bioinspired therapeutics that induces homeostatic renormalization, here we show that the collection of molecules released from four cell types, including mesenchymal stem cells, fibroblast, neural stem cells, and astrocytes, rescues degenerating neurons and cells. Specifically, oxidative stress induced in a human recombinant TDP-43- or FUS-tGFP U2OS cell line by exposure to sodium arsenite was shown to be significantly reduced by our collection of molecules using in vitro imaging of FUS and TDP-43 stress granules. Furthermore, we also show that the collective secretome rescues cortical neurons from glutamate toxicity as evidenced by increased neurite outgrowth, reduced LDH release, and reduced caspase 3/7 activity. These data are the first in a series supporting the development of stem cell-based exosome systems therapeutics that uses a physiological renormalization strategy to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Greg Maguire
- BioRegenerative Sciences, Inc.San DiegoCalifornia
- Auditory Sound Waves, LLCSan DiegoCalifornia
| | - Lee Paler
- BioRegenerative Sciences, Inc.San DiegoCalifornia
- Auditory Sound Waves, LLCSan DiegoCalifornia
| | - Linda Green
- BioRegenerative Sciences, Inc.San DiegoCalifornia
| | | | | | | |
Collapse
|
130
|
PAX3 Confers Functional Heterogeneity in Skeletal Muscle Stem Cell Responses to Environmental Stress. Cell Stem Cell 2019; 24:958-973.e9. [PMID: 31006622 DOI: 10.1016/j.stem.2019.03.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/04/2019] [Accepted: 03/22/2019] [Indexed: 11/22/2022]
Abstract
Muscle satellite cells (MuSCs) are the quiescent muscle stem cells required for adult skeletal muscle repair. The impact of environmental stress such as pollution on MuSC behavior remains unexplored. We evaluated the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure, a ubiquitous and highly toxic pollutant, on MuSCs by combining in vivo mouse molecular genetic models with ex vivo studies. While all MuSCs express the transcription factor PAX7, we show that a subset also express PAX3 and exhibit resistance to environmental stress. Upon systemic TCDD treatment, PAX3-negative MuSCs display impaired survival, atypical activation, and sporadic differentiation through xenobiotic aryl hydrocarbon receptor signaling. We further show that PAX3-positive MuSCs become sensitized to environmental stress when PAX3 function is impaired and that PAX3-mediated induction of mTORC1 is required for protection. Our study, therefore, identifies a functional heterogeneity of MuSCs in response to environmental stress controlled by PAX3.
Collapse
|
131
|
Dell'Orso S, Juan AH, Ko KD, Naz F, Perovanovic J, Gutierrez-Cruz G, Feng X, Sartorelli V. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions. Development 2019; 146:dev.174177. [PMID: 30890574 DOI: 10.1242/dev.174177] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/07/2019] [Indexed: 12/15/2022]
Abstract
Dedicated stem cells ensure postnatal growth, repair and homeostasis of skeletal muscle. Following injury, muscle stem cells (MuSCs) exit from quiescence and divide to reconstitute the stem cell pool and give rise to muscle progenitors. The transcriptomes of pooled MuSCs have provided a rich source of information for describing the genetic programs of distinct static cell states; however, bulk microarray and RNA sequencing provide only averaged gene expression profiles, blurring the heterogeneity and developmental dynamics of asynchronous MuSC populations. Instead, the granularity required to identify distinct cell types, states, and their dynamics can be afforded by single cell analysis. We were able to compare the transcriptomes of thousands of MuSCs and primary myoblasts isolated from homeostatic or regenerating muscles by single cell RNA sequencing. Using computational approaches, we could reconstruct dynamic trajectories and place, in a pseudotemporal manner, the transcriptomes of individual MuSC within these trajectories. This approach allowed for the identification of distinct clusters of MuSCs and primary myoblasts with partially overlapping but distinct transcriptional signatures, as well as the description of metabolic pathways associated with defined MuSC states.
Collapse
Affiliation(s)
- Stefania Dell'Orso
- Genome Technology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA stefania.dell'
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Kyung-Dae Ko
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Faiza Naz
- Genome Technology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Jelena Perovanovic
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Gustavo Gutierrez-Cruz
- Genome Technology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Xuesong Feng
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA stefania.dell'
| |
Collapse
|
132
|
Forcina L, Miano C, Pelosi L, Musarò A. An Overview about the Biology of Skeletal Muscle Satellite Cells. Curr Genomics 2019; 20:24-37. [PMID: 31015789 PMCID: PMC6446479 DOI: 10.2174/1389202920666190116094736] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 12/14/2022] Open
Abstract
The peculiar ability of skeletal muscle tissue to operate adaptive changes during post-natal de-velopment and adulthood has been associated with the existence of adult somatic stem cells. Satellite cells, occupying an exclusive niche within the adult muscle tissue, are considered bona fide stem cells with both stem-like properties and myogenic activities. Indeed, satellite cells retain the capability to both maintain the quiescence in uninjured muscles and to be promptly activated in response to growth or re-generative signals, re-engaging the cell cycle. Activated cells can undergo myogenic differentiation or self-renewal moving back to the quiescent state. Satellite cells behavior and their fate decision are finely controlled by mechanisms involving both cell-autonomous and external stimuli. Alterations in these regu-latory networks profoundly affect muscle homeostasis and the dynamic response to tissue damage, con-tributing to the decline of skeletal muscle that occurs under physio-pathologic conditions. Although the clear myogenic activity of satellite cells has been described and their pivotal role in muscle growth and regeneration has been reported, a comprehensive picture of inter-related mechanisms guiding muscle stem cell activity has still to be defined. Here, we reviewed the main regulatory networks determining satellite cell behavior. In particular, we focused on genetic and epigenetic mechanisms underlining satel-lite cell maintenance and commitment. Besides intrinsic regulations, we reported current evidences about the influence of environmental stimuli, derived from other cell populations within muscle tissue, on satel-lite cell biology.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Laura Pelosi
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14 Rome 00161, Italy
| |
Collapse
|
133
|
Abstract
Stem cells can reside in a state of reversible growth arrest, or quiescence, for prolonged periods of time. Although quiescence has long been viewed as a dormant, low-activity state, increasing evidence suggests that quiescence represents states of poised potential and active restraint, as stem cells "idle" in anticipation of activation, proliferation, and differentiation. Improved understanding of quiescent stem cell dynamics is leading to novel approaches to enhance maintenance and repair of aged or diseased tissues. In this Review, we discuss recent advances in our understanding of stem cell quiescence and techniques enabling more refined analyses of quiescence in vivo.
Collapse
Affiliation(s)
- Cindy T J van Velthoven
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
134
|
Sagot I, Laporte D. The cell biology of quiescent yeast – a diversity of individual scenarios. J Cell Sci 2019; 132:132/1/jcs213025. [DOI: 10.1242/jcs.213025] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT
Most cells, from unicellular to complex organisms, spend part of their life in quiescence, a temporary non-proliferating state. Although central for a variety of essential processes including tissue homeostasis, development and aging, quiescence is poorly understood. In fact, quiescence encompasses various cellular situations depending on the cell type and the environmental niche. Quiescent cell properties also evolve with time, adding another layer of complexity. Studying quiescence is, above all, limited by the fact that a quiescent cell can be recognized as such only after having proved that it is capable of re-proliferating. Recent cellular biology studies in yeast have reported the relocalization of hundreds of proteins and the reorganization of several cellular machineries upon proliferation cessation. These works have revealed that quiescent cells can display various properties, shedding light on a plethora of individual behaviors. The deciphering of the molecular mechanisms beyond these reorganizations, together with the understanding of their cellular functions, have begun to provide insights into the physiology of quiescent cells. In this Review, we discuss recent findings and emerging concepts in Saccharomyces cerevisiae quiescent cell biology.
Collapse
Affiliation(s)
- Isabelle Sagot
- Centre National de la Recherche Scientifique, Université de Bordeaux-Institut de Biochimie et Génétique Cellulaires, UMR5095-33077 Bordeaux cedex, France
| | - Damien Laporte
- Centre National de la Recherche Scientifique, Université de Bordeaux-Institut de Biochimie et Génétique Cellulaires, UMR5095-33077 Bordeaux cedex, France
| |
Collapse
|
135
|
Evano B, Tajbakhsh S. Skeletal muscle stem cells in comfort and stress. NPJ Regen Med 2018; 3:24. [PMID: 30588332 PMCID: PMC6303387 DOI: 10.1038/s41536-018-0062-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
Investigations on developmental and regenerative myogenesis have led to major advances in decrypting stem cell properties and potential, as well as their interactions within the evolving niche. As a consequence, regenerative myogenesis has provided a forum to investigate intrinsic regulators of stem cell properties as well as extrinsic factors, including stromal cells, during normal growth and following injury and disease. Here we review some of the latest advances in the field that have exposed fundamental processes including regulation of stress following trauma and ageing, senescence, DNA damage control and modes of symmetric and asymmetric cell divisions. Recent studies have begun to explore the nature of the niche that is distinct in different muscle groups, and that is altered from prenatal to postnatal stages, and during ageing. We also discuss heterogeneities among muscle stem cells and how distinct properties within the quiescent and proliferating cell states might impact on homoeostasis and regeneration. Interestingly, cellular quiescence, which was thought to be a passive cell state, is regulated by multiple mechanisms, many of which are deregulated in various contexts including ageing. These and other factors including metabolic activity and genetic background can impact on the efficiency of muscle regeneration.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
136
|
Sahu A, Mamiya H, Shinde SN, Cheikhi A, Winter LL, Vo NV, Stolz D, Roginskaya V, Tang WY, St Croix C, Sanders LH, Franti M, Van Houten B, Rando TA, Barchowsky A, Ambrosio F. Age-related declines in α-Klotho drive progenitor cell mitochondrial dysfunction and impaired muscle regeneration. Nat Commun 2018; 9:4859. [PMID: 30451844 PMCID: PMC6242898 DOI: 10.1038/s41467-018-07253-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 10/23/2018] [Indexed: 01/15/2023] Open
Abstract
While young muscle is capable of restoring the original architecture of damaged myofibers, aged muscle displays a markedly reduced regeneration. We show that expression of the "anti-aging" protein, α-Klotho, is up-regulated within young injured muscle as a result of transient Klotho promoter demethylation. However, epigenetic control of the Klotho promoter is lost with aging. Genetic inhibition of α-Klotho in vivo disrupted muscle progenitor cell (MPC) lineage progression and impaired myofiber regeneration, revealing a critical role for α-Klotho in the regenerative cascade. Genetic silencing of Klotho in young MPCs drove mitochondrial DNA (mtDNA) damage and decreased cellular bioenergetics. Conversely, supplementation with α-Klotho restored mtDNA integrity and bioenergetics of aged MPCs to youthful levels in vitro and enhanced functional regeneration of aged muscle in vivo in a temporally-dependent manner. These studies identify a role for α-Klotho in the regulation of MPC mitochondrial function and implicate α-Klotho declines as a driver of impaired muscle regeneration with age.
Collapse
MESH Headings
- Aging/genetics
- Aging/metabolism
- Aging/pathology
- Animals
- DNA Methylation
- DNA, Mitochondrial/genetics
- DNA, Mitochondrial/metabolism
- Epigenesis, Genetic
- Gene Expression Regulation, Developmental
- Glucuronidase
- Klotho Proteins
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/genetics
- Mitochondria/metabolism
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myoblasts/metabolism
- Myoblasts/pathology
- Promoter Regions, Genetic
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Regeneration/genetics
- Signal Transduction
- Stem Cells/metabolism
- Stem Cells/pathology
Collapse
Affiliation(s)
- A Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213, PA, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - H Mamiya
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, 15260, PA, USA
| | - S N Shinde
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213, PA, USA
| | - A Cheikhi
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213, PA, USA
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, 15213, PA, USA
| | - L L Winter
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, 15260, PA, USA
| | - N V Vo
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, 15213, PA, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - D Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - V Roginskaya
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, 15232, PA, USA
| | - W Y Tang
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, 21218-2608, MD, USA
| | - C St Croix
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, 15261, PA, USA
| | - L H Sanders
- Department of Neurology, Duke University School of Medicine, Durham, 27704, NC, USA
| | - M Franti
- Research Beyond Borders: Boehringer-Ingelheim Pharmaceuticals, Ridgefield, 06877, CT, USA
| | - B Van Houten
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, 15232, PA, USA
| | - T A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
- The Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Center for Tissue Regeneration, Restoration and Repair, Veterans Affairs Hospital, Palo Alto, CA, 94036, USA
| | - A Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, 15261, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, 15232, PA, USA
| | - F Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, 15213, PA, USA.
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, 15261, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, 15260, PA, USA.
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, 15213, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, 15219, PA, USA.
| |
Collapse
|
137
|
Verma M, Asakura Y, Murakonda BSR, Pengo T, Latroche C, Chazaud B, McLoon LK, Asakura A. Muscle Satellite Cell Cross-Talk with a Vascular Niche Maintains Quiescence via VEGF and Notch Signaling. Cell Stem Cell 2018; 23:530-543.e9. [PMID: 30290177 PMCID: PMC6178221 DOI: 10.1016/j.stem.2018.09.007] [Citation(s) in RCA: 217] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/19/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022]
Abstract
Skeletal muscle is a complex tissue containing tissue resident muscle stem cells (satellite cells) (MuSCs) important for postnatal muscle growth and regeneration. Quantitative analysis of the biological function of MuSCs and the molecular pathways responsible for a potential juxtavascular niche for MuSCs is currently lacking. We utilized fluorescent reporter mice and muscle tissue clearing to investigate the proximity of MuSCs to capillaries in 3 dimensions. We show that MuSCs express abundant VEGFA, which recruits endothelial cells (ECs) in vitro, whereas blocking VEGFA using both a vascular endothelial growth factor (VEGF) inhibitor and MuSC-specific VEGFA gene deletion reduces the proximity of MuSCs to capillaries. Importantly, this proximity to the blood vessels was associated with MuSC self-renewal in which the EC-derived Notch ligand Dll4 induces quiescence in MuSCs. We hypothesize that MuSCs recruit capillary ECs via VEGFA, and in return, ECs maintain MuSC quiescence though Dll4.
Collapse
Affiliation(s)
- Mayank Verma
- Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Bhavani Sai Rohit Murakonda
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Thomas Pengo
- University of Minnesota Informatics Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Claire Latroche
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | | | - Linda K McLoon
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Ophthalmology and Visual Neurosciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN, USA; Paul & Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN, USA; Department of Neurology, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
138
|
Khodabukus A, Prabhu N, Wang J, Bursac N. In Vitro Tissue-Engineered Skeletal Muscle Models for Studying Muscle Physiology and Disease. Adv Healthc Mater 2018; 7:e1701498. [PMID: 29696831 PMCID: PMC6105407 DOI: 10.1002/adhm.201701498] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/18/2018] [Indexed: 12/18/2022]
Abstract
Healthy skeletal muscle possesses the extraordinary ability to regenerate in response to small-scale injuries; however, this self-repair capacity becomes overwhelmed with aging, genetic myopathies, and large muscle loss. The failure of small animal models to accurately replicate human muscle disease, injury and to predict clinically-relevant drug responses has driven the development of high fidelity in vitro skeletal muscle models. Herein, the progress made and challenges ahead in engineering biomimetic human skeletal muscle tissues that can recapitulate muscle development, genetic diseases, regeneration, and drug response is discussed. Bioengineering approaches used to improve engineered muscle structure and function as well as the functionality of satellite cells to allow modeling muscle regeneration in vitro are also highlighted. Next, a historical overview on the generation of skeletal muscle cells and tissues from human pluripotent stem cells, and a discussion on the potential of these approaches to model and treat genetic diseases such as Duchenne muscular dystrophy, is provided. Finally, the need to integrate multiorgan microphysiological systems to generate improved drug discovery technologies with the potential to complement or supersede current preclinical animal models of muscle disease is described.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Neel Prabhu
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Jason Wang
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| | - Nenad Bursac
- Department of Biomedical Engineering Duke University 101 Science Drive, FCIEMAS 1427, Durham, NC 27708-90281, USA
| |
Collapse
|
139
|
Machado L, Esteves de Lima J, Fabre O, Proux C, Legendre R, Szegedi A, Varet H, Ingerslev LR, Barrès R, Relaix F, Mourikis P. In Situ Fixation Redefines Quiescence and Early Activation of Skeletal Muscle Stem Cells. Cell Rep 2018; 21:1982-1993. [PMID: 29141227 DOI: 10.1016/j.celrep.2017.10.080] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/13/2017] [Accepted: 10/21/2017] [Indexed: 12/12/2022] Open
Abstract
State of the art techniques have been developed to isolate and analyze cells from various tissues, aiming to capture their in vivo state. However, the majority of cell isolation protocols involve lengthy mechanical and enzymatic dissociation steps followed by flow cytometry, exposing cells to stress and disrupting their physiological niche. Focusing on adult skeletal muscle stem cells, we have developed a protocol that circumvents the impact of isolation procedures and captures cells in their native quiescent state. We show that current isolation protocols induce major transcriptional changes accompanied by specific histone modifications while having negligible effects on DNA methylation. In addition to proposing a protocol to avoid isolation-induced artifacts, our study reveals previously undetected quiescence and early activation genes of potential biological interest.
Collapse
Affiliation(s)
- Léo Machado
- Biology of the Neuromuscular System, INSERM IMRB U955-E10, UPEC, ENVA, EFS, Creteil 94000, France
| | - Joana Esteves de Lima
- Biology of the Neuromuscular System, INSERM IMRB U955-E10, UPEC, ENVA, EFS, Creteil 94000, France
| | - Odile Fabre
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Proux
- Institut Pasteur, Plate-forme Transcriptome & Epigenome, Biomics, Centre d'Innovation et Recherche Technologique (Citech), Paris, France
| | - Rachel Legendre
- Institut Pasteur, Plate-forme Transcriptome & Epigenome, Biomics, Centre d'Innovation et Recherche Technologique (Citech), Paris, France; Institut Pasteur, Hub Bioinformatique et Biostatistique, Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI, USR 3756 IP CNRS), Paris, France
| | - Anikó Szegedi
- Biology of the Neuromuscular System, INSERM IMRB U955-E10, UPEC, ENVA, EFS, Creteil 94000, France
| | - Hugo Varet
- Institut Pasteur, Plate-forme Transcriptome & Epigenome, Biomics, Centre d'Innovation et Recherche Technologique (Citech), Paris, France; Institut Pasteur, Hub Bioinformatique et Biostatistique, Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI, USR 3756 IP CNRS), Paris, France
| | - Lars Roed Ingerslev
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frédéric Relaix
- Biology of the Neuromuscular System, INSERM IMRB U955-E10, UPEC, ENVA, EFS, Creteil 94000, France.
| | - Philippos Mourikis
- Biology of the Neuromuscular System, INSERM IMRB U955-E10, UPEC, ENVA, EFS, Creteil 94000, France
| |
Collapse
|
140
|
Díaz-Carballo D, Saka S, Klein J, Rennkamp T, Acikelli AH, Malak S, Jastrow H, Wennemuth G, Tempfer C, Schmitz I, Tannapfel A, Strumberg D. A Distinct Oncogenerative Multinucleated Cancer Cell Serves as a Source of Stemness and Tumor Heterogeneity. Cancer Res 2018; 78:2318-2331. [PMID: 29440172 DOI: 10.1158/0008-5472.can-17-1861] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/20/2017] [Accepted: 02/06/2018] [Indexed: 11/16/2022]
Abstract
The effects of anticancer treatments on cell heterogeneity and their proliferative potential play an important role in tumor persistence and metastasis. However, little is known about de-polyploidization, cell fate, and physiologic stemness of the resulting cell populations. Here, we describe a distinctive cell type termed "pregnant" P1 cells found within chemotherapy-refractory ovarian tumors, which generate and gestate daughter generation Gn cells intracytoplasmically. Release of Gn cells occurred by ejection through crevices in the P1 cell membrane by body contractions or using a funiculus-like structure. These events characterized a not yet described mechanism of cell segregation. Maternal P1 cells were principally capable of surviving parturition events and continued to breed and nurture Gn progenies. In addition, P1 cells were competent to horizontally transmit offspring Gn cells into other specific proximal cells, injecting them to receptor R1 cells via cell-cell tunneling. This process represents a new mechanism used by tumor cells to invade surrounding tissues and ensure life cycles. In contrast to the pregnant P1 cells with low expression of stem cell markers despite their physiologic stemness, the first offspring generations of daughter G1 cells expressed high levels of ovarian cancer stem cell markers. Furthermore, both P1 and Gn cells overexpressed multiple human endogenous retroviral envelope proteins. Moreover, programmed death-ligand 1 and the immunosuppressive domain of the retroviral envelope proteins were also overexpressed in P1 cells, suggesting effective protection against the host immune system. Together, our data suggest that P1 oncogenerative cancer cells exhibit a not yet described cell biological mechanism of persistence and transmission of malignant cells in patients with advanced cancers.Significance: P1 oncogenerative cell entities express low levels of CSC markers, which are characteristic of their histological origin. Cancer Res; 78(9); 2318-31. ©2018 AACR.
Collapse
Affiliation(s)
- David Díaz-Carballo
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany.
| | - Sahitya Saka
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| | - Jacqueline Klein
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| | - Tobias Rennkamp
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| | - Ali H Acikelli
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| | - Sascha Malak
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| | - Holger Jastrow
- Institute of Anatomy, University of Duisburg-Essen, Medical School, Essen, Germany
| | - Gunther Wennemuth
- Institute of Anatomy, University of Duisburg-Essen, Medical School, Essen, Germany
| | - Clemens Tempfer
- Gynaecology and Obstetrics, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| | - Inge Schmitz
- Institute of Pathology, Ruhr University Bochum, Medical School, Bochum, Germany
| | - Andrea Tannapfel
- Institute of Pathology, Ruhr University Bochum, Medical School, Bochum, Germany
| | - Dirk Strumberg
- Institute of Molecular Oncology and Experimental Therapeutics. Division of Haematology and Oncology, Marienhospital Herne, Ruhr University Bochum, Medical School, Herne, Germany
| |
Collapse
|
141
|
Relaix F, Machado L. Waking up muscle stem cells: PI3K signalling is ringing. EMBO J 2018; 37:embj.201899297. [PMID: 29581098 DOI: 10.15252/embj.201899297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Frederic Relaix
- Inserm, IMRB U955-E10, Créteil, France.,Faculté de Médecine, Université Paris Est Créteil, Créteil, France.,Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.,Etablissement Français du Sang, Créteil, France.,DHU Pepsy& Centre de Référence des Maladies Neuromusculaires GNMH, APHP, Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Léo Machado
- Inserm, IMRB U955-E10, Créteil, France.,Faculté de Médecine, Université Paris Est Créteil, Créteil, France.,Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| |
Collapse
|
142
|
Wang G, Zhu H, Situ C, Han L, Yu Y, Cheung TH, Liu K, Wu Z. p110α of PI3K is necessary and sufficient for quiescence exit in adult muscle satellite cells. EMBO J 2018; 37:embj.201798239. [PMID: 29581096 DOI: 10.15252/embj.201798239] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/19/2018] [Accepted: 02/23/2018] [Indexed: 11/09/2022] Open
Abstract
Adult mouse muscle satellite cells (MuSCs) are quiescent in uninjured muscles. Upon injury, MuSCs exit quiescence in vivo to become activated, re-enter the cell cycle to proliferate, and differentiate to repair the damaged muscles. It remains unclear which extrinsic cues and intrinsic signaling pathways regulate quiescence exit during MuSC activation. Here, we demonstrated that inducible MuSC-specific deletion of p110α, a catalytic subunit of phosphatidylinositol 3-kinase (PI3K), rendered MuSCs unable to exit quiescence, resulting in severely impaired MuSC proliferation and muscle regeneration. Genetic reactivation of mTORC1, or knockdown of FoxOs, in p110α-null MuSCs partially rescued the above defects, making them key effectors downstream of PI3K in regulating quiescence exit. c-Jun was found to be a key transcriptional target of the PI3K/mTORC1 signaling axis essential for MuSC quiescence exit. Moreover, induction of a constitutively active PI3K in quiescent MuSCs resulted in spontaneous MuSC activation in uninjured muscles and subsequent depletion of the MuSC pool. Thus, PI3K-p110α is both necessary and sufficient for MuSCs to exit quiescence in response to activating signals.
Collapse
Affiliation(s)
- Gang Wang
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Han Zhu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Chenghao Situ
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Lifang Han
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Youqian Yu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Kai Liu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| | - Zhenguo Wu
- Division of Life Science, Center for Stem Cell Research, Center for Systems Biology and Human Health, the State Key Laboratory in Neuroscience, Hong Kong University of Science & Technology, Kowloon Hong Kong, China
| |
Collapse
|