1
|
Fancello I, Willett S, Castiglioni C, Amer S, Santoleri S, Bragg L, Galli F, Cossu G. TNAP expressing adventitial pericytes contribute to myogenesis during foetal development. Vascul Pharmacol 2025; 159:107489. [PMID: 40097085 DOI: 10.1016/j.vph.2025.107489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE During growth and differentiation of skeletal muscle, cell types other than canonical myoblasts can be recruited to a myogenic fate. Among these, TNAP+ pericytes can differentiate into skeletal or smooth muscle cells during postnatal growth and contribute to muscle regeneration. However, their role in muscle development has not been investigated. This study aims to characterise pericyte fate choices during embryonic and foetal myogenesis, occurring in the second half of gestation. APPROACH AND RESULTS Using Cre-loxP lineage tracing with multiple reporters including the multifluorescent Confetti, we labelled TNAP+ precursors in vivo and assessed the smooth or skeletal muscle differentiation in their lineage at a perinatal stage. We found that TNAP+ cells contribute in vivo to skeletal and smooth muscle cells, as well as other pericytes, also during pre-natal muscle development. The resulting clones showed that such fate choices are likely to depend on distinct unipotent progenitors rather than multipotent progenitors. In addition, we isolated and differentiated in vitro foetal cells derived from TNAP+ precursors, which showed that they are not spontaneously myogenic unless co-cultured with other skeletal muscle cells. CONCLUSIONS This work extends our understanding of the differentiative potency of these non- canonical skeletal muscle progenitors during prenatal life, with a view to a future application of this knowledge to optimise cell therapies for muscle wasting disorders.
Collapse
Affiliation(s)
- I Fancello
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Willett
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - C Castiglioni
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Amer
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - S Santoleri
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - L Bragg
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - F Galli
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK
| | - G Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, FBMH, University of Manchester, UK; Institute of Experimental Neurology, Division of Neurosciences, Ospedale San Raffaele, Milan, Italy; Experimental and Clinical Research Center, Charité Medical Faculty, Max Delbrück Center Berlin, Germany.
| |
Collapse
|
2
|
Girolamo DD, Benavente-Diaz M, Murolo M, Grimaldi A, Lopes PT, Evano B, Kuriki M, Gioftsidi S, Laville V, Tinevez JY, Letort G, Mella S, Tajbakhsh S, Comai G. Extraocular muscle stem cells exhibit distinct cellular properties associated with non-muscle molecular signatures. Development 2024; 151:dev202144. [PMID: 38240380 DOI: 10.1242/dev.202144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/27/2023] [Indexed: 02/22/2024]
Abstract
Skeletal muscle stem cells (MuSCs) are recognised as functionally heterogeneous. Cranial MuSCs are reported to have greater proliferative and regenerative capacity when compared with those in the limb. A comprehensive understanding of the mechanisms underlying this functional heterogeneity is lacking. Here, we have used clonal analysis, live imaging and single cell transcriptomic analysis to identify crucial features that distinguish extraocular muscle (EOM) from limb muscle stem cell populations. A MyogeninntdTom reporter showed that the increased proliferation capacity of EOM MuSCs correlates with deferred differentiation and lower expression of the myogenic commitment gene Myod. Unexpectedly, EOM MuSCs activated in vitro expressed a large array of extracellular matrix components typical of mesenchymal non-muscle cells. Computational analysis underscored a distinct co-regulatory module, which is absent in limb MuSCs, as driver of these features. The EOM transcription factor network, with Foxc1 as key player, appears to be hardwired to EOM identity as it persists during growth, disease and in vitro after several passages. Our findings shed light on how high-performing MuSCs regulate myogenic commitment by remodelling their local environment and adopting properties not generally associated with myogenic cells.
Collapse
Affiliation(s)
- Daniela Di Girolamo
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Maria Benavente-Diaz
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005 Paris, France
| | - Melania Murolo
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Alexandre Grimaldi
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005 Paris, France
| | - Priscilla Thomas Lopes
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Brendan Evano
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Mao Kuriki
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Stamatia Gioftsidi
- Université Paris-Est, 77420 Champs-sur- Marne, France
- Freie Universität Berlin, 14195 Berlin, Germany
- Inserm, IMRB U955-E10, 94000 Créteil, France
| | - Vincent Laville
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Jean-Yves Tinevez
- Institut Pasteur, Université Paris Cité, Image Analysis Hub, 75015 Paris, France
| | - Gaëlle Letort
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Université de Paris Cité, CNRS UMR 3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Sebastian Mella
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Glenda Comai
- Stem Cells and Development Unit, 25 rue du Dr Roux, Institut Pasteur, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| |
Collapse
|
3
|
Xu D, Wan B, Qiu K, Wang Y, Zhang X, Jiao N, Yan E, Wu J, Yu R, Gao S, Du M, Liu C, Li M, Fan G, Yin J. Single-Cell RNA-Sequencing Provides Insight into Skeletal Muscle Evolution during the Selection of Muscle Characteristics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305080. [PMID: 37870215 PMCID: PMC10724408 DOI: 10.1002/advs.202305080] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/27/2023] [Indexed: 10/24/2023]
Abstract
Skeletal muscle comprises a large, heterogeneous assortment of cell populations that interact to maintain muscle homeostasis, but little is known about the mechanism that controls myogenic development in response to artificial selection. Different pig (Sus scrofa) breeds exhibit distinct muscle phenotypes resulting from domestication and selective breeding. Using unbiased single-cell transcriptomic sequencing analysis (scRNA-seq), the impact of artificial selection on cell profiles is investigated in neonatal skeletal muscle of pigs. This work provides panoramic muscle-resident cell profiles and identifies novel and breed-specific cells, mapping them on pseudotime trajectories. Artificial selection has elicited significant changes in muscle-resident cell profiles, while conserving signs of generational environmental challenges. These results suggest that fibro-adipogenic progenitors serve as a cellular interaction hub and that specific transcription factors identified here may serve as candidate target regulons for the pursuit of a specific muscle phenotype. Furthermore, a cross-species comparison of humans, mice, and pigs illustrates the conservation and divergence of mammalian muscle ontology. The findings of this study reveal shifts in cellular heterogeneity, novel cell subpopulations, and their interactions that may greatly facilitate the understanding of the mechanism underlying divergent muscle phenotypes arising from artificial selection.
Collapse
Affiliation(s)
- Doudou Xu
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Boyang Wan
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Kai Qiu
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Yubo Wang
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Xin Zhang
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
- Molecular Design Breeding Frontier Science Center of the Ministry of EducationBeijingChina
| | - Ning Jiao
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Enfa Yan
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Jiangwei Wu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYangling712100China
| | - Run Yu
- Beijing National Day SchoolBeijing100039China
| | - Shuai Gao
- Key Laboratory of Animal GeneticsCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
| | - Min Du
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciences and School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
| | | | - Mingzhou Li
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengdu625014China
| | - Guoping Fan
- Department of Human GeneticsDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition and feedingCollege of Animal Science and TechnologyChina Agricultural UniversityBeijing100193China
- Molecular Design Breeding Frontier Science Center of the Ministry of EducationBeijingChina
| |
Collapse
|
4
|
Abbas H, Olivere LA, Padgett ME, Schmidt CA, Gilmore BF, McCord TJ, Southerland KW, McClung JM, Kontos CD. Muscle progenitor cells are required for skeletal muscle regeneration and prevention of adipogenesis after limb ischemia. Front Cardiovasc Med 2023; 10:1118738. [PMID: 36937923 PMCID: PMC10017542 DOI: 10.3389/fcvm.2023.1118738] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Skeletal muscle injury in peripheral artery disease (PAD) has been attributed to vascular insufficiency, however evidence has demonstrated that muscle cell responses play a role in determining outcomes in limb ischemia. Here, we demonstrate that genetic ablation of Pax7+ muscle progenitor cells (MPCs) in a model of hindlimb ischemia (HLI) inhibited muscle regeneration following ischemic injury, despite a lack of morphological or physiological changes in resting muscle. Compared to control mice (Pax7WT), the ischemic limb of Pax7-deficient mice (Pax7Δ) was unable to generate significant force 7 or 28 days after HLI. A significant increase in adipose was observed in the ischemic limb 28 days after HLI in Pax7Δ mice, which replaced functional muscle. Adipogenesis in Pax7Δ mice corresponded with a significant increase in PDGFRα+ fibro/adipogenic progenitors (FAPs). Inhibition of FAPs with batimastat decreased muscle adipose but increased fibrosis. In vitro, Pax7Δ MPCs failed to form myotubes but displayed increased adipogenesis. Skeletal muscle from patients with critical limb threatening ischemia displayed increased adipose in more ischemic regions of muscle, which corresponded with fewer satellite cells. Collectively, these data demonstrate that Pax7+ MPCs are required for muscle regeneration after ischemia and suggest that muscle regeneration may be an important therapeutic target in PAD.
Collapse
Affiliation(s)
- Hasan Abbas
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | | | - Michael E. Padgett
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | - Cameron A. Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Brian F. Gilmore
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Timothy J. McCord
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin W. Southerland
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Joseph M. McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Heart Institute, East Carolina University, Greenville, NC, United States
| | - Christopher D. Kontos
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
- Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
5
|
Choi S, Ferrari G, Moyle LA, Mackinlay K, Naouar N, Jalal S, Benedetti S, Wells C, Muntoni F, Tedesco FS. Assessing and enhancing migration of human myogenic progenitors using directed iPS cell differentiation and advanced tissue modelling. EMBO Mol Med 2022; 14:e14526. [PMID: 36161772 PMCID: PMC9549733 DOI: 10.15252/emmm.202114526] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
Muscle satellite stem cells (MuSCs) are responsible for skeletal muscle growth and regeneration. Despite their differentiation potential, human MuSCs have limited in vitro expansion and in vivo migration capacity, limiting their use in cell therapies for diseases affecting multiple skeletal muscles. Several protocols have been developed to derive MuSC-like progenitors from human induced pluripotent stem (iPS) cells (hiPSCs) to establish a source of myogenic cells with controllable proliferation and differentiation. However, current hiPSC myogenic derivatives also suffer from limitations of cell migration, ultimately delaying their clinical translation. Here we use a multi-disciplinary approach including bioinformatics and tissue engineering to show that DLL4 and PDGF-BB improve migration of hiPSC-derived myogenic progenitors. Transcriptomic analyses demonstrate that this property is conserved across species and multiple hiPSC lines, consistent with results from single cell motility profiling. Treated cells showed enhanced trans-endothelial migration in transwell assays. Finally, increased motility was detected in a novel humanised assay to study cell migration using 3D artificial muscles, harnessing advanced tissue modelling to move hiPSCs closer to future muscle gene and cell therapies.
Collapse
Affiliation(s)
- SungWoo Choi
- The Francis Crick InstituteLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Giulia Ferrari
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Louise A Moyle
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
- Present address:
Institute of Biomedical EngineeringUniversity of TorontoTorontoONCanada
| | - Kirsty Mackinlay
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
- Present address:
Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Naira Naouar
- Institut de Biologie Paris Seine FR3631, Plateforme de Bioinformatique ARTbioSorbonne UniversitéParisFrance
| | - Salma Jalal
- The Francis Crick InstituteLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Sara Benedetti
- UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Christine Wells
- Centre for Stem Cell SystemsThe University of MelbourneMelbourneVICAustralia
| | - Francesco Muntoni
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research CentreLondonUK
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for ChildrenLondonUK
| | - Francesco Saverio Tedesco
- The Francis Crick InstituteLondonUK
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
- Dubowitz Neuromuscular CentreUCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for ChildrenLondonUK
| |
Collapse
|
6
|
Liu C, Niu K, Xiao Q. Updated perspectives on vascular cell specification and pluripotent stem cell-derived vascular organoids for studying vasculopathies. Cardiovasc Res 2022; 118:97-114. [PMID: 33135070 PMCID: PMC8752356 DOI: 10.1093/cvr/cvaa313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Vasculopathy is a pathological process occurring in the blood vessel wall, which could affect the haemostasis and physiological functions of all the vital tissues/organs and is one of the main underlying causes for a variety of human diseases including cardiovascular diseases. Current pharmacological interventions aiming to either delay or stop progression of vasculopathies are suboptimal, thus searching novel, targeted, risk-reducing therapeutic agents, or vascular grafts with full regenerative potential for patients with vascular abnormalities are urgently needed. Since first reported, pluripotent stem cells (PSCs), particularly human-induced PSCs, have open new avenue in all research disciplines including cardiovascular regenerative medicine and disease remodelling. Assisting with recent technological breakthroughs in tissue engineering, in vitro construction of tissue organoid made a tremendous stride in the past decade. In this review, we provide an update of the main signal pathways involved in vascular cell differentiation from human PSCs and an extensive overview of PSC-derived tissue organoids, highlighting the most recent discoveries in the field of blood vessel organoids as well as vascularization of other complex tissue organoids, with the aim of discussing the key cellular and molecular players in generating vascular organoids.
Collapse
MESH Headings
- Blood Vessels/metabolism
- Blood Vessels/pathology
- Blood Vessels/physiopathology
- Cell Culture Techniques
- Cell Differentiation
- Cell Lineage
- Cells, Cultured
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Humans
- Induced Pluripotent Stem Cells/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Organoids
- Phenotype
- Signal Transduction
- Vascular Diseases/metabolism
- Vascular Diseases/pathology
- Vascular Diseases/physiopathology
Collapse
Affiliation(s)
- Chenxin Liu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Kaiyuan Niu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases at The Second Affiliated Hospital
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou, Guangdong 511436, China
| |
Collapse
|
7
|
Cossu G, Tonlorenzi R, Brunelli S, Sampaolesi M, Messina G, Azzoni E, Benedetti S, Biressi S, Bonfanti C, Bragg L, Camps J, Cappellari O, Cassano M, Ciceri F, Coletta M, Covarello D, Crippa S, Cusella-De Angelis MG, De Angelis L, Dellavalle A, Diaz-Manera J, Galli D, Galli F, Gargioli C, Gerli MFM, Giacomazzi G, Galvez BG, Hoshiya H, Guttinger M, Innocenzi A, Minasi MG, Perani L, Previtali SC, Quattrocelli M, Ragazzi M, Roostalu U, Rossi G, Scardigli R, Sirabella D, Tedesco FS, Torrente Y, Ugarte G. Mesoangioblasts at 20: From the embryonic aorta to the patient bed. Front Genet 2022; 13:1056114. [PMID: 36685855 PMCID: PMC9845585 DOI: 10.3389/fgene.2022.1056114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/31/2022] [Indexed: 01/06/2023] Open
Abstract
In 2002 we published an article describing a population of vessel-associated progenitors that we termed mesoangioblasts (MABs). During the past decade evidence had accumulated that during muscle development and regeneration things may be more complex than a simple sequence of binary choices (e.g., dorsal vs. ventral somite). LacZ expressing fibroblasts could fuse with unlabelled myoblasts but not among themselves or with other cell types. Bone marrow derived, circulating progenitors were able to participate in muscle regeneration, though in very small percentage. Searching for the embryonic origin of these progenitors, we identified them as originating at least in part from the embryonic aorta and, at later stages, from the microvasculature of skeletal muscle. While continuing to investigate origin and fate of MABs, the fact that they could be expanded in vitro (also from human muscle) and cross the vessel wall, suggested a protocol for the cell therapy of muscular dystrophies. We tested this protocol in mice and dogs before proceeding to the first clinical trial on Duchenne Muscular Dystrophy patients that showed safety but minimal efficacy. In the last years, we have worked to overcome the problem of low engraftment and tried to understand their role as auxiliary myogenic progenitors during development and regeneration.
Collapse
Affiliation(s)
- Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine. University of Manchester, Manchester, United Kingdom
- Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
- Muscle Research Unit, Charité Medical Faculty and Max Delbrück Center, Berlin, Germany
- *Correspondence: Giulio Cossu, ; Rossana Tonlorenzi, ; Silvia Brunelli, ; Maurilio Sampaolesi, ; Graziella Messina,
| | - Rossana Tonlorenzi
- Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
- *Correspondence: Giulio Cossu, ; Rossana Tonlorenzi, ; Silvia Brunelli, ; Maurilio Sampaolesi, ; Graziella Messina,
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
- *Correspondence: Giulio Cossu, ; Rossana Tonlorenzi, ; Silvia Brunelli, ; Maurilio Sampaolesi, ; Graziella Messina,
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Histology and Medical Embryology Unit, Department of Anatomy, Forensic Medicine and Orthopaedics, Sapienza University, Rome, Italy
- *Correspondence: Giulio Cossu, ; Rossana Tonlorenzi, ; Silvia Brunelli, ; Maurilio Sampaolesi, ; Graziella Messina,
| | - Graziella Messina
- Department of Biosciences, University of Milan, Milan, Italy
- *Correspondence: Giulio Cossu, ; Rossana Tonlorenzi, ; Silvia Brunelli, ; Maurilio Sampaolesi, ; Graziella Messina,
| | - Emanuele Azzoni
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Sara Benedetti
- UCL Great Ormond Street Institute of Child Health and NIHR GOSH Biomedical Research Centre, London, United Kingdom
| | - Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO) and Dulbecco Telethon Institute, University of Trento, Trento, Italy
| | - Chiara Bonfanti
- Department of Biosciences, University of Milan, Milan, Italy
| | - Laricia Bragg
- Division of Cell Matrix Biology and Regenerative Medicine. University of Manchester, Manchester, United Kingdom
| | - Jordi Camps
- Bayer AG, Research and Development, Pharmaceuticals, Berlin, Germany
| | - Ornella Cappellari
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, Bari, Italy
| | | | - Fabio Ciceri
- Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marcello Coletta
- Histology and Medical Embryology Unit, Department of Anatomy, Forensic Medicine and Orthopaedics, Sapienza University, Rome, Italy
| | | | - Stefania Crippa
- San Raffaele-Telethon Institute of Gene Theray, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Luciana De Angelis
- Histology and Medical Embryology Unit, Department of Anatomy, Forensic Medicine and Orthopaedics, Sapienza University, Rome, Italy
| | | | - Jordi Diaz-Manera
- John Walton Muscular Dystrophy Research Centre, Newcastle University, United Kingdom
| | - Daniela Galli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Francesco Galli
- Division of Cell Matrix Biology and Regenerative Medicine. University of Manchester, Manchester, United Kingdom
| | - Cesare Gargioli
- Department of Biology, University of Tor Vergata, Rome, Italy
| | - Mattia F. M. Gerli
- UCL Department of Surgical Biotechnology and Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Beatriz G. Galvez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Universidad Complutense de Madrid, Madrid, Spain
| | | | | | - Anna Innocenzi
- Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - M. Giulia Minasi
- Lavitaminasi, Clinical Nutrition and Reproductive Medicine, Rome, Italy
| | - Laura Perani
- Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, University of Cincinnati, Cincinnati, OH, United States
| | | | - Urmas Roostalu
- Roche Institute for Translational Bioengineering (ITB), pRED Basel, Basel, Switzerland
| | - Giuliana Rossi
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Raffaella Scardigli
- Columbia Stem Cell Initiative, Department of Rehabilitation and Regenerative Medicine, Columbia University, New York, United States
| | - Dario Sirabella
- University College London, Great Ormond Street Hospital for Children and the Francis Crick Institute, London, United Kingdom
| | - Francesco Saverio Tedesco
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Yvan Torrente
- UCL Great Ormond Street Institute of Child Health and NIHR GOSH Biomedical Research Centre, London, United Kingdom
| | - Gonzalo Ugarte
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| |
Collapse
|
8
|
Mele A, Mantuano P, Fonzino A, Rana F, Capogrosso RF, Sanarica F, Rolland JF, Cappellari O, De Luca A. Ultrasonography validation for early alteration of diaphragm echodensity and function in the mdx mouse model of Duchenne muscular dystrophy. PLoS One 2021; 16:e0245397. [PMID: 33434240 PMCID: PMC7802948 DOI: 10.1371/journal.pone.0245397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 12/29/2020] [Indexed: 12/26/2022] Open
Abstract
The mdx mouse model of Duchenne muscular dystrophy is characterized by functional and structural alterations of the diaphragm since early stages of pathology, closely resembling patients' condition. In recent years, ultrasonography has been proposed as a useful longitudinal non-invasive technique to assess mdx diaphragm dysfunction and evaluate drug efficacy over time. To date, only a few preclinical studies have been conducted. Therefore, an independent validation of this method by different laboratories is needed to increase results reliability and reduce biases. Here, we performed diaphragm ultrasonography in 3- and 6-month-old mdx mice, the preferred age-window for pharmacology studies. The alteration of diaphragm function over time was measured as diaphragm ultrasound movement amplitude. At the same time points, a first-time assessment of diaphragm echodensity was performed, as an experimental index of progressive loss of contractile tissue. A parallel evaluation of other in vivo and ex vivo dystrophy-relevant readouts was carried out. Both 3- and 6-month-old mdx mice showed a significant decrease in diaphragm amplitude compared to wild type (wt) mice. This index was well-correlated either with in vivo running performance or ex vivo isometric tetanic force of isolated diaphragm. In addition, diaphragms from 6-month-old dystrophic mice were also highly susceptible to eccentric contraction ex vivo. Importantly, we disclosed an age-dependent increase in echodensity in mdx mice not observed in wt animals, which was independent from abdominal wall thickness. This was accompanied by a notable increase of pro-fibrotic TGF-β1 levels in the mdx diaphragm and of non-muscle tissue amount in diaphragm sections stained by hematoxylin & eosin. Our findings corroborate the usefulness of diaphragm ultrasonography in preclinical drug studies as a powerful tool to monitor mdx pathology progression since early stages.
Collapse
Affiliation(s)
- Antonietta Mele
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Paola Mantuano
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Adriano Fonzino
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Rana
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | | | - Francesca Sanarica
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | | | - Ornella Cappellari
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Annamaria De Luca
- Section of Pharmacology, Department of Pharmacy—Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
9
|
Choi S, Ferrari G, Tedesco FS. Cellular dynamics of myogenic cell migration: molecular mechanisms and implications for skeletal muscle cell therapies. EMBO Mol Med 2020; 12:e12357. [PMID: 33210465 PMCID: PMC7721365 DOI: 10.15252/emmm.202012357] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/02/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Directional cell migration is a critical process underlying morphogenesis and post-natal tissue regeneration. During embryonic myogenesis, migration of skeletal myogenic progenitors is essential to generate the anlagen of limbs, diaphragm and tongue, whereas in post-natal skeletal muscles, migration of muscle satellite (stem) cells towards regions of injury is necessary for repair and regeneration of muscle fibres. Additionally, safe and efficient migration of transplanted cells is critical in cell therapies, both allogeneic and autologous. Although various myogenic cell types have been administered intramuscularly or intravascularly, functional restoration has not been achieved yet in patients with degenerative diseases affecting multiple large muscles. One of the key reasons for this negative outcome is the limited migration of donor cells, which hinders the overall cell engraftment potential. Here, we review mechanisms of myogenic stem/progenitor cell migration during skeletal muscle development and post-natal regeneration. Furthermore, strategies utilised to improve migratory capacity of myogenic cells are examined in order to identify potential treatments that may be applied to future transplantation protocols.
Collapse
Affiliation(s)
- SungWoo Choi
- Department of Cell and Developmental Biology, University College London, London, UK.,The Francis Crick Institute, London, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, UK.,The Francis Crick Institute, London, UK.,Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
10
|
Anguiano T, Sahu A, Qian B, Tang WY, Ambrosio F, Barchowsky A. Arsenic Directs Stem Cell Fate by Imparting Notch Signaling Into the Extracellular Matrix Niche. Toxicol Sci 2020; 177:494-505. [PMID: 32647881 DOI: 10.1093/toxsci/kfaa106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Compromise of skeletal muscle metabolism and composition may underlie the etiology of cardiovascular and metabolic disease risk from environmental arsenic exposures. We reported that arsenic impairs muscle maintenance and regeneration by inducing maladaptive mitochondrial phenotypes in muscle stem cells (MuSC), connective tissue fibroblasts (CTF), and myofibers. We also found that arsenic imparts a dysfunctional memory in the extracellular matrix (ECM) that disrupts the MuSC niche and is sufficient to favor the expansion and differentiation of fibrogenic MuSC subpopulations. To investigate the signaling mechanisms involved in imparting a dysfunctional ECM, we isolated skeletal muscle tissue and CTF from mice exposed to 0 or 100 μg/l arsenic in their drinking water for 5 weeks. ECM elaborated by arsenic-exposed CTF decreased myogenesis and increased fibrogenic/adipogenic MuSC subpopulations and differentiation. However, treating arsenic-exposed mice with SS-31, a mitochondrially targeted peptide that repairs the respiratory chain, reversed the arsenic-promoted CTF phenotype to one that elaborated an ECM supporting normal myogenic differentiation. SS-31 treatment also reversed arsenic-induced Notch1 expression, resulting in an improved muscle regeneration after injury. We found that persistent arsenic-induced CTF Notch1 expression caused the elaboration of dysfunctional ECM with increased expression of the Notch ligand DLL4. This DLL4 in the ECM was responsible for misdirecting MuSC myogenic differentiation. These data indicate that arsenic impairs muscle maintenance and regenerative capacity by targeting CTF mitochondria and mitochondrially directed expression of dysfunctional regulators in the stem cell niche. Therapies that restore muscle cell mitochondria may effectively treat arsenic-induced skeletal muscle dysfunction and compositional decline.
Collapse
Affiliation(s)
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation
| | - Baoli Qian
- Department of Environmental and Occupational Health
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health
| | - Fabrisia Ambrosio
- Department of Environmental and Occupational Health.,Department of Physical Medicine and Rehabilitation.,McGowan Institute for Regenerative Medicine.,Department of Bioengineering
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
11
|
Gerli MFM, Moyle LA, Benedetti S, Ferrari G, Ucuncu E, Ragazzi M, Constantinou C, Louca I, Sakai H, Ala P, De Coppi P, Tajbakhsh S, Cossu G, Tedesco FS. Combined Notch and PDGF Signaling Enhances Migration and Expression of Stem Cell Markers while Inducing Perivascular Cell Features in Muscle Satellite Cells. Stem Cell Reports 2019; 12:461-473. [PMID: 30745033 PMCID: PMC6409426 DOI: 10.1016/j.stemcr.2019.01.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/31/2022] Open
Abstract
Satellite cells are responsible for skeletal muscle regeneration. Upon activation, they proliferate as transient amplifying myoblasts, most of which fuse into regenerating myofibers. Despite their remarkable differentiation potential, these cells have limited migration capacity, which curtails clinical use for widespread forms of muscular dystrophy. Conversely, skeletal muscle perivascular cells have less myogenic potential but better migration capacity than satellite cells. Here we show that modulation of Notch and PDGF pathways, involved in developmental specification of pericytes, induces perivascular cell features in adult mouse and human satellite cell-derived myoblasts. DLL4 and PDGF-BB-treated cells express markers of perivascular cells and associate with endothelial networks while also upregulating markers of satellite cell self-renewal. Moreover, treated cells acquire trans-endothelial migration ability while remaining capable of engrafting skeletal muscle upon intramuscular transplantation. These results extend our understanding of muscle stem cell fate plasticity and provide a druggable pathway with clinical relevance for muscle cell therapy.
Collapse
Affiliation(s)
- Mattia Francesco Maria Gerli
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK; Stem Cell and Regenerative Medicine Section, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
| | - Louise Anne Moyle
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK
| | - Sara Benedetti
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK; Molecular and Cellular Immunology Section, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, WC1N 1EH London, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK
| | - Ekin Ucuncu
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK
| | - Chrystalla Constantinou
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK
| | - Irene Louca
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK
| | - Hiroshi Sakai
- Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France; CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Pierpaolo Ala
- The Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK
| | - Shahragim Tajbakhsh
- Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France; CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, M13 9PL Manchester, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, WC1E 6DE London, UK; The Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, UK.
| |
Collapse
|
12
|
Ando K, Wang W, Peng D, Chiba A, Lagendijk AK, Barske L, Crump JG, Stainier DYR, Lendahl U, Koltowska K, Hogan BM, Fukuhara S, Mochizuki N, Betsholtz C. Peri-arterial specification of vascular mural cells from naïve mesenchyme requires Notch signaling. Development 2019; 146:dev.165589. [PMID: 30642834 DOI: 10.1242/dev.165589] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022]
Abstract
Mural cells (MCs) are essential for blood vessel stability and function; however, the mechanisms that regulate MC development remain incompletely understood, in particular those involved in MC specification. Here, we investigated the first steps of MC formation in zebrafish using transgenic reporters. Using pdgfrb and abcc9 reporters, we show that the onset of expression of abcc9, a pericyte marker in adult mice and zebrafish, occurs almost coincidentally with an increment in pdgfrb expression in peri-arterial mesenchymal cells, suggesting that these transcriptional changes mark the specification of MC lineage cells from naïve pdgfrb low mesenchymal cells. The emergence of peri-arterial pdgfrb high MCs required Notch signaling. We found that pdgfrb-positive cells express notch2 in addition to notch3, and although depletion of notch2 or notch3 failed to block MC emergence, embryos depleted of both notch2 and notch3 lost mesoderm- as well as neural crest-derived pdgfrb high MCs. Using reporters that read out Notch signaling and Notch2 receptor cleavage, we show that Notch activation in the mesenchyme precedes specification into pdgfrb high MCs. Taken together, these results show that Notch signaling is necessary for peri-arterial MC specification.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden .,Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan
| | - Weili Wang
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan
| | - Anne K Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Lindsey Barske
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 77 Stockholm, Sweden.,Department of Medicine, Huddinge, Karolinska Institutet, Integrated Cardio Metabolic Centre (ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden
| | - Katarzyna Koltowska
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden.,Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Shigetomo Fukuhara
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan.,Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School Musashi Kosugi Hospital, Kawasaki, Kanagawa 211 8533, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan.,AMED-CREST, Department of Cell Biology, National Cerebral and Cardiovascular Center, 5-7-1, Suita, Osaka 565 8565, Japan
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden.,Department of Medicine, Huddinge, Karolinska Institutet, Integrated Cardio Metabolic Centre (ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden
| |
Collapse
|
13
|
Moyle LA, Tedesco FS, Benedetti S. Pericytes in Muscular Dystrophies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:319-344. [PMID: 31147885 DOI: 10.1007/978-3-030-16908-4_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The muscular dystrophies are an heterogeneous group of inherited myopathies characterised by the progressive wasting of skeletal muscle tissue. Pericytes have been shown to make muscle in vitro and to contribute to skeletal muscle regeneration in several animal models, although recent data has shown this to be controversial. In fact, some pericyte subpopulations have been shown to contribute to fibrosis and adipose deposition in muscle. In this chapter, we explore the identity and the multifaceted role of pericytes in dystrophic muscle, potential therapeutic applications and the current need to overcome the hurdles of characterisation (both to identify pericyte subpopulations and track cell fate), to prevent deleterious differentiation towards myogenic-inhibiting subpopulations, and to improve cell proliferation and engraftment efficacy.
Collapse
Affiliation(s)
- Louise Anne Moyle
- Institute of Biomaterials and Biomedical Engineering, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, UK.
- Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Sara Benedetti
- Great Ormond Street Institute of Child Health, University College London, London, UK.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
14
|
Lee LL, Chintalgattu V. Pericytes in the Heart. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:187-210. [PMID: 30937870 DOI: 10.1007/978-3-030-11093-2_11] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mural cells known as pericytes envelop the endothelial layer of microvessels throughout the body and have been described to have tissue-specific functions. Cardiac pericytes are abundantly found in the heart, but they are relatively understudied. Currently, their importance is emerging in cardiovascular homeostasis and dysfunction due to their pleiotropism. They are known to play key roles in vascular tone and vascular integrity as well as angiogenesis. However, their dysfunctional presence and/or absence is critical in the mechanisms that lead to cardiac pathologies such as myocardial infarction, fibrosis, and thrombosis. Moreover, they are targeted as a therapeutic potential due to their mesenchymal properties that could allow them to repair and regenerate a damaged heart. They are also sought after as a cell-based therapy based on their healing potential in preclinical studies of animal models of myocardial infarction. Therefore, recognizing the importance of cardiac pericytes and understanding their biology will lead to new therapeutic concepts.
Collapse
Affiliation(s)
- Linda L Lee
- Department of CardioMetabolic Disorders, Amgen Research and Discovery, Amgen Inc., South San Francisco, CA, USA
| | - Vishnu Chintalgattu
- Department of CardioMetabolic Disorders, Amgen Research and Discovery, Amgen Inc., South San Francisco, CA, USA.
| |
Collapse
|
15
|
Lu J, Liu QH, Wang F, Tan JJ, Deng YQ, Peng XH, Liu X, Zhang B, Xu X, Li XP. Exosomal miR-9 inhibits angiogenesis by targeting MDK and regulating PDK/AKT pathway in nasopharyngeal carcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:147. [PMID: 30001734 PMCID: PMC6044044 DOI: 10.1186/s13046-018-0814-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 06/26/2018] [Indexed: 01/11/2023]
Abstract
Background Exosomes are small vesicles containing a wide range of functional proteins, mRNA and miRNA. Exosomal miRNAs from cancer cells play crucial roles in mediating cell-cell communication and tumor-microenvironment cross talk, specifically in enabling metastasis and promoting angiogenesis. We focused on miR-9 that was identified as a tumor suppressor previously in nasopharyngeal carcinoma (NPC) tumorigenesis. Methods Differential centrifugation, transmission electron microscopy and nanoparticle tracking analysis were used to isolate and identify exosomes. Quantitative PCR and western blotting analysis were used to detect miR-9, pri-miR-9, CD63, TSG101, MDK, P70S6K P-Ser424 and PDK1 P-Ser241 expression. Laser confocal microscopy was used to trace exosomal miR-9 secreted by NPC cells into HUVECs. The effect of exosomal miR-9 on cell migration and tube formation of HUVECs in vivo and vitro was assessed by using migration assay, tube formation assay and matrigel plug assay, respectively. Bioinformatics analysis and luciferase reporter assay were utilized to confirm the binding of exosomal miR-9 to the 3′untranslated region (3′-UTR) of MDK, while Phosphorylation Array was performed to identify AKT Pathway in HUVECs treated with exosomal miR-9. Furthermore, Immunohistochemistry (IHC) and in situ hybridization (ISH) was used to detected miR-9, CD31 and MDK expression in human NPC tumor samples. Results NPC cells transfected with miR-9-overexpressing lentivirus, released miR-9 in exosomes. Exosomal miR-9 directly suppressed its target gene - MDK in endothelial cells. Mechanistic analyses revealed that exosomal miR-9 from NPC cells inhibited endothelial tube formation and migration by targeting MDK and regulating PDK/AKT signaling pathway. Additionally, the level of MDK was upregulated in NPC tumor samples and was positively correlated with microvessel density. Notably, the level of exosomal miR-9 was positively correlated with overall survival, and MDK overexpression was positively associated with poor prognosis in NPC patients, suggesting the clinical relevance and prognostic value of exosomal miR-9 and MDK. Conclusions Taken together, our data identify an extracellular anti-angiogenic role for tumor-derived, exosome-associated miR-9 in NPC tumorigenesis and prompt further investigation into exosome-based therapies for cancer treatment. Electronic supplementary material The online version of this article (10.1186/s13046-018-0814-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juan Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi-Hui Liu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Wang
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jia-Jie Tan
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Hong Peng
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiong Liu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bao Zhang
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Xia Xu
- Department of Otolaryngology-Head and Neck Surgery, Guangzhou General Hospital of People's Liberation Army of China, Guangzhou, China
| | - Xiang-Ping Li
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
16
|
Naldaiz‐Gastesi N, Bahri OA, López de Munain A, McCullagh KJA, Izeta A. The panniculus carnosus muscle: an evolutionary enigma at the intersection of distinct research fields. J Anat 2018; 233:275-288. [PMID: 29893024 PMCID: PMC6081499 DOI: 10.1111/joa.12840] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2018] [Indexed: 12/13/2022] Open
Abstract
The panniculus carnosus is a thin striated muscular layer intimately attached to the skin and fascia of most mammals, where it provides skin twitching and contraction functions. In humans, the panniculus carnosus is conserved at sparse anatomical locations with high interindividual variability, and it is considered of no functional significance (most possibly being a remnant of evolution). Diverse research fields (such as anatomy, dermatology, myology, neuroscience, surgery, veterinary science) use this unique muscle as a model, but several unknowns and misconceptions remain in the literature. In this article, we review what is currently known about panniculus carnosus structure, development, anatomical location, response to environmental stimuli and potential function(s), with the aim of putting together the evidence arising from the different research communities and raising interest in this unique muscle, which we postulate as an ideal model for both vascular and muscular research.
Collapse
Affiliation(s)
- Neia Naldaiz‐Gastesi
- Tissue Engineering GroupBioengineering AreaInstituto BiodonostiaSan SebastianSpain
- Neuroscience AreaInstituto BiodonostiaSan SebastianSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
| | - Ola A. Bahri
- Department of PhysiologyHuman Biology BuildingSchool of MedicineNational University of Ireland GalwayGalwayIreland
- Regenerative Medicine InstituteNational University of Ireland GalwayGalwayIreland
| | - Adolfo López de Munain
- Neuroscience AreaInstituto BiodonostiaSan SebastianSpain
- CIBERNED, Instituto de Salud Carlos IIIMadridSpain
- Faculty of Medicine and DentistryUPV‐EHUSan SebastianSpain
- Department of NeurologyHospital Universitario DonostiaSan SebastianSpain
| | - Karl J. A. McCullagh
- Department of PhysiologyHuman Biology BuildingSchool of MedicineNational University of Ireland GalwayGalwayIreland
- Regenerative Medicine InstituteNational University of Ireland GalwayGalwayIreland
| | - Ander Izeta
- Tissue Engineering GroupBioengineering AreaInstituto BiodonostiaSan SebastianSpain
- Department of Biomedical EngineeringSchool of EngineeringTecnun‐University of NavarraSan SebastianSpain
| |
Collapse
|
17
|
Sinha S, Santoro MM. New models to study vascular mural cell embryonic origin: implications in vascular diseases. Cardiovasc Res 2018; 114:481-491. [PMID: 29385541 DOI: 10.1093/cvr/cvy005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/23/2018] [Indexed: 02/15/2024] Open
Abstract
A key question in vascular biology is how the diversity of origin of vascular mural cells, namely smooth muscle cells (SMCs) and pericytes influences vessel properties, in particular the regional propensity to vascular diseases. This review therefore first describes the role and regulation of mural cells during vascular formation, with a focus on embryonic origin. We then consider the evidence that connects heterogeneities in SMC and pericyte origins with disease. Since this idea has major implications for understanding and modelling human disease, then there is a pressing need for new model systems to investigate mural cell development and the consequences of heterogeneity. Recent advances arising from in vitro strategies for deriving mural cells from human pluripotent stem cells as well as from the zebrafish model will be discussed and the medical relevance of these discoveries will be highlighted.
Collapse
Affiliation(s)
- Sanjay Sinha
- Anne McLaren Laboratory, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute, Forvie Site, University of Cambridge, Robinson Way, Cambridge CB2 0SZ, UK
- Department of Medicine, Addenbrookes Hospital, Box 157, Hills Rd, Cambridge, CB2 0QQ, UK
| | - Massimo Mattia Santoro
- Laboratory of Angiogenesis and Redox Metabolism, Department of Biology, University of Padua, 35131 Padova, Italy
| |
Collapse
|
18
|
Nandagopal N, Santat LA, LeBon L, Sprinzak D, Bronner ME, Elowitz MB. Dynamic Ligand Discrimination in the Notch Signaling Pathway. Cell 2018; 172:869-880.e19. [PMID: 29398116 PMCID: PMC6414217 DOI: 10.1016/j.cell.2018.01.002] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 08/10/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023]
Abstract
The Notch signaling pathway comprises multiple ligands that are used in distinct biological contexts. In principle, different ligands could activate distinct target programs in signal-receiving cells, but it is unclear how such ligand discrimination could occur. Here, we show that cells use dynamics to discriminate signaling by the ligands Dll1 and Dll4 through the Notch1 receptor. Quantitative single-cell imaging revealed that Dll1 activates Notch1 in discrete, frequency-modulated pulses that specifically upregulate the Notch target gene Hes1. By contrast, Dll4 activates Notch1 in a sustained, amplitude-modulated manner that predominantly upregulates Hey1 and HeyL. Ectopic expression of Dll1 or Dll4 in chick neural crest produced opposite effects on myogenic differentiation, showing that ligand discrimination can occur in vivo. Finally, analysis of chimeric ligands suggests that ligand-receptor clustering underlies dynamic encoding of ligand identity. The ability of the pathway to utilize ligands as distinct communication channels has implications for diverse Notch-dependent processes.
Collapse
Affiliation(s)
- Nagarajan Nandagopal
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Leah A Santat
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Lauren LeBon
- Calico Life Sciences, 1170 Veterans Boulevard, South San Francisco, CA 94080, USA
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Howard Hughes Medical Institute, Division of Biology and Biological Engineering, Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
19
|
Bigas A, Porcheri C. Notch and Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:235-263. [DOI: 10.1007/978-3-319-89512-3_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Sommese L, Zullo A, Schiano C, Mancini FP, Napoli C. Possible Muscle Repair in the Human Cardiovascular System. Stem Cell Rev Rep 2017; 13:170-191. [PMID: 28058671 DOI: 10.1007/s12015-016-9711-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The regenerative potential of tissues and organs could promote survival, extended lifespan and healthy life in multicellular organisms. Niches of adult stemness are widely distributed and lead to the anatomical and functional regeneration of the damaged organ. Conversely, muscular regeneration in mammals, and humans in particular, is very limited and not a single piece of muscle can fully regrow after a severe injury. Therefore, muscle repair after myocardial infarction is still a chimera. Recently, it has been recognized that epigenetics could play a role in tissue regrowth since it guarantees the maintenance of cellular identity in differentiated cells and, therefore, the stability of organs and tissues. The removal of these locks can shift a specific cell identity back to the stem-like one. Given the gradual loss of tissue renewal potential in the course of evolution, in the last few years many different attempts to retrieve such potential by means of cell therapy approaches have been performed in experimental models. Here we review pathways and mechanisms involved in the in vivo repair of cardiovascular muscle tissues in humans. Moreover, we address the ongoing research on mammalian cardiac muscle repair based on adult stem cell transplantation and pro-regenerative factor delivery. This latter issue, involving genetic manipulations of adult cells, paves the way for developing possible therapeutic strategies in the field of cardiovascular muscle repair.
Collapse
Affiliation(s)
- Linda Sommese
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.
| | - Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy.,CEINGE Advanced Biotechnologies, s.c.ar.l, Naples, Italy
| | | | - Francesco P Mancini
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Claudio Napoli
- Department of Internal and Specialty Medicine, U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology, Azienda Ospedaliera Universitaria, Università degli Studi della Campania "Luigi Vanvitelli", Piazza Miraglia 2, 80138, Naples, Italy.,IRCCS Foundation SDN, Naples, Italy
| |
Collapse
|
21
|
Demolli S, Doddaballapur A, Devraj K, Stark K, Manavski Y, Eckart A, Zehendner CM, Lucas T, Korff T, Hecker M, Massberg S, Liebner S, Kaluza D, Boon RA, Dimmeler S. Shear stress-regulated miR-27b controls pericyte recruitment by repressing SEMA6A and SEMA6D. Cardiovasc Res 2017; 113:681-691. [PMID: 28453731 DOI: 10.1093/cvr/cvx032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/22/2017] [Indexed: 11/14/2022] Open
Abstract
AIMS Vessel maturation involves the recruitment of mural cells such as pericytes and smooth muscle cells. Laminar shear stress is a major trigger for vessel maturation, but the molecular mechanisms by which shear stress affects recruitment of pericytes are unclear. MicroRNAs (miRs) are small non-coding RNAs, which post-transcriptionally control gene expression. The aim of the present study was to unveil the mechanism by which shear stress-regulated microRNAs contribute to vessel maturation. METHODS AND RESULTS Here, we show that laminar shear stress increased miR-27a and miR-27b expression in vitro and in ex vivo in mouse femoral artery explants. Overexpression of miR-27b in endothelial cells increased pericyte adhesion and pericyte recruitment in vitro. In vitro barrier function of endothelial-pericyte co-cultures was augmented by miR-27b overexpression, whereas inhibition of miR-27a/b reduced adhesion and pericyte coverage and decreased barrier functions. In vivo, pharmacological inhibition of miR-27a/b by locked nucleic acid antisense oligonucleotides significantly reduced pericyte coverage and increased water content in the murine uterus. MiR-27b overexpression repressed semaphorins (SEMA), which mediate repulsive signals, and the vessel destabilizing human but not mouse Angiopoietin-2 (Ang-2). Silencing of SEMA6A and SEMA6D rescued the reduced pericyte adhesion by miR-27 inhibition. Furthermore, inhibition of SEMA6D increased barrier function of an endothelial-pericyte co-culture in vitro. CONCLUSION The present study demonstrates for the first time that shear stress-regulated miR-27b promotes the interaction of endothelial cells with pericytes, partly by repressing SEMA6A and SEMA6D.
Collapse
Affiliation(s)
- Shemsi Demolli
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Anuradha Doddaballapur
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Kavi Devraj
- Institute for Neurology (Edinger Institute), Goethe University, 60528 Frankfurt, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Yosif Manavski
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Annekathrin Eckart
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Christoph M Zehendner
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
- ZIM III, Department of Cardiology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Tina Lucas
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, 69120 Heidelberg, Germany
| | - Markus Hecker
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, 69120 Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Munich, Germany
| | - Stefan Liebner
- Institute for Neurology (Edinger Institute), Goethe University, 60528 Frankfurt, Germany
| | - David Kaluza
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Reinier A Boon
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite RheinMain, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite RheinMain, Germany
| |
Collapse
|
22
|
Miller SR, Perera SN, Baker CVH. Constitutively active Notch1 converts cranial neural crest-derived frontonasal mesenchyme to perivascular cells in vivo. Biol Open 2017; 6:317-325. [PMID: 28183698 PMCID: PMC5374403 DOI: 10.1242/bio.023887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Perivascular/mural cells originate from either the mesoderm or the cranial neural crest. Regardless of their origin, Notch signalling is necessary for their formation. Furthermore, in both chicken and mouse, constitutive Notch1 activation (via expression of the Notch1 intracellular domain) is sufficient in vivo to convert trunk mesoderm-derived somite cells to perivascular cells, at the expense of skeletal muscle. In experiments originally designed to investigate the effect of premature Notch1 activation on the development of neural crest-derived olfactory ensheathing glial cells (OECs), we used in ovo electroporation to insert a tetracycline-inducible NotchΔE construct (encoding a constitutively active mutant of mouse Notch1) into the genome of chicken cranial neural crest cell precursors, and activated NotchΔE expression by doxycycline injection at embryonic day 4. NotchΔE-targeted cells formed perivascular cells within the frontonasal mesenchyme, and expressed a perivascular marker on the olfactory nerve. Hence, constitutively activating Notch1 is sufficient in vivo to drive not only somite cells, but also neural crest-derived frontonasal mesenchyme and perhaps developing OECs, to a perivascular cell fate. These results also highlight the plasticity of neural crest-derived mesenchyme and glia. Summary: Sustained Notch1 activation is sufficient to drive cranial neural crest-derived frontonasal mesenchyme to adopt a perivascular (mural) cell fate in developing chick embryos.
Collapse
Affiliation(s)
- Sophie R Miller
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Surangi N Perera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Clare V H Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
23
|
Currie JD, Kawaguchi A, Traspas RM, Schuez M, Chara O, Tanaka EM. Live Imaging of Axolotl Digit Regeneration Reveals Spatiotemporal Choreography of Diverse Connective Tissue Progenitor Pools. Dev Cell 2016; 39:411-423. [PMID: 27840105 PMCID: PMC5127896 DOI: 10.1016/j.devcel.2016.10.013] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 01/05/2023]
Abstract
Connective tissues-skeleton, dermis, pericytes, fascia-are a key cell source for regenerating the patterned skeleton during axolotl appendage regeneration. This complexity has made it difficult to identify the cells that regenerate skeletal tissue. Inability to identify these cells has impeded a mechanistic understanding of blastema formation. By tracing cells during digit tip regeneration using brainbow transgenic axolotls, we show that cells from each connective tissue compartment have distinct spatial and temporal profiles of proliferation, migration, and differentiation. Chondrocytes proliferate but do not migrate into the regenerate. In contrast, pericytes proliferate, then migrate into the blastema and give rise solely to pericytes. Periskeletal cells and fibroblasts contribute the bulk of digit blastema cells and acquire diverse fates according to successive waves of migration that choreograph their proximal-distal and tissue contributions. We further show that platelet-derived growth factor signaling is a potent inducer of fibroblast migration, which is required to form the blastema.
Collapse
Affiliation(s)
- Joshua D Currie
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauserstrasse 108, 01307 Dresden, Germany.
| | - Akane Kawaguchi
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Ricardo Moreno Traspas
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Maritta Schuez
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany
| | - Osvaldo Chara
- Center for Information Services and High Performance Computing (ZIH), Technische Universität Dresden, 01062 Dresden, Germany; Systems Biology Group (SysBio), Instituto de Física de Líquidos y Sistemas Biológicos (IFLySIB), CONICET, Universidad Nacional de La Plata (UNLP), B1900BTE La Plata, Buenos Aires, Argentina
| | - Elly M Tanaka
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstrasse 105, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauserstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
24
|
Birbrair A, Sattiraju A, Zhu D, Zulato G, Batista I, Nguyen VT, Messi ML, Solingapuram Sai KK, Marini FC, Delbono O, Mintz A. Novel Peripherally Derived Neural-Like Stem Cells as Therapeutic Carriers for Treating Glioblastomas. Stem Cells Transl Med 2016; 6:471-481. [PMID: 28191774 PMCID: PMC5442817 DOI: 10.5966/sctm.2016-0007] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 08/09/2016] [Indexed: 12/29/2022] Open
Abstract
Glioblastoma (GBM), an aggressive grade IV astrocytoma, is the most common primary malignant adult brain tumor characterized by extensive invasiveness, heterogeneity, and angiogenesis. Standard treatment options such as radiation and chemotherapy have proven to be only marginally effective in treating GBM because of its invasive nature. Therefore, extensive efforts have been put forth to develop tumor‐tropic stem cells as viable therapeutic vehicles with potential to treat even the most invasive tumor cells that are harbored within areas of normal brain. To this end, we discovered a newly described NG2‐expressing cell that we isolated from a distinct pericyte subtype found abundantly in cultures derived from peripheral muscle. In this work, we show the translational significance of these peripherally derived neural‐like stem cells (NLSC) and their potential to migrate toward tumors and act as therapeutic carriers. We demonstrate that these NLSCs exhibit in vitro and in vivo GBM tropism. Furthermore, NLSCs did not promote angiogenesis or transform into tumor‐associated stromal cells, which are concerns raised when using other common stem cells, such as mesenchymal stem cells and induced neural stem cells, as therapeutic carriers. We also demonstrate the potential of NLSCs to express a prototype therapeutic, tumor necrosis factor α‐related apoptosis‐inducing ligand and kill GBM cells in vitro. These data demonstrate the therapeutic potential of our newly characterized NLSC against GBM. Stem Cells Translational Medicine2017;6:471–481
Collapse
Affiliation(s)
- Alexander Birbrair
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Internal Medicine‐Gerontology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
- Department of Pathology, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Anirudh Sattiraju
- Department of Radiology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
- Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Winston‐Salem, North Carolina, USA
| | - Dongqin Zhu
- Department of Radiology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
| | - Gilberto Zulato
- Department of Radiology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
| | - Izadora Batista
- Department of Radiology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
| | - Van T. Nguyen
- Department of Radiology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
| | - Maria Laura Messi
- Department of Internal Medicine‐Gerontology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
| | - Kiran Kumar Solingapuram Sai
- Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Winston‐Salem, North Carolina, USA
| | - Frank C. Marini
- Wake Forest Institute for Regenerative Medicine, Winston‐Salem, North Carolina, USA
| | - Osvaldo Delbono
- Department of Internal Medicine‐Gerontology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston‐Salem, North Carolina, USA
- Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Winston‐Salem, North Carolina, USA
| |
Collapse
|
25
|
Murray IR, Baily JE, Chen WCW, Dar A, Gonzalez ZN, Jensen AR, Petrigliano FA, Deb A, Henderson NC. Skeletal and cardiac muscle pericytes: Functions and therapeutic potential. Pharmacol Ther 2016; 171:65-74. [PMID: 27595928 DOI: 10.1016/j.pharmthera.2016.09.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pericytes are periendothelial mesenchymal cells residing within the microvasculature. Skeletal muscle and cardiac pericytes are now recognized to fulfill an increasing number of functions in normal tissue homeostasis, including contributing to microvascular function by maintaining vessel stability and regulating capillary flow. In the setting of muscle injury, pericytes contribute to a regenerative microenvironment through release of trophic factors and by modulating local immune responses. In skeletal muscle, pericytes also directly enhance tissue healing by differentiating into myofibers. Conversely, pericytes have also been implicated in the development of disease states, including fibrosis, heterotopic ossication and calcification, atherosclerosis, and tumor angiogenesis. Despite increased recognition of pericyte heterogeneity, it is not yet clear whether specific subsets of pericytes are responsible for individual functions in skeletal and cardiac muscle homeostasis and disease.
Collapse
Affiliation(s)
- Iain R Murray
- BHF Center for Vascular Regeneration and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK; Department of Trauma and Orthopaedic Surgery, The University of Edinburgh, Edinburgh, UK
| | - James E Baily
- BHF Center for Vascular Regeneration and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - William C W Chen
- Reseach Laboratory of Electronics and Department of Biological Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Ayelet Dar
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Zaniah N Gonzalez
- BHF Center for Vascular Regeneration and MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Andrew R Jensen
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Frank A Petrigliano
- Orthopedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine & Molecular Cell and Developmental Biology, and Eli and Edythe Broad Institute of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, CA, USA.
| | - Neil C Henderson
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
26
|
Baeten JT, Lilly B. Notch Signaling in Vascular Smooth Muscle Cells. ADVANCES IN PHARMACOLOGY 2016; 78:351-382. [PMID: 28212801 DOI: 10.1016/bs.apha.2016.07.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The Notch signaling pathway is a highly conserved pathway involved in cell fate determination in embryonic development and also functions in the regulation of physiological processes in several systems. It plays an especially important role in vascular development and physiology by influencing angiogenesis, vessel patterning, arterial/venous specification, and vascular smooth muscle biology. Aberrant or dysregulated Notch signaling is the cause of or a contributing factor to many vascular disorders, including inherited vascular diseases, such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, associated with degeneration of the smooth muscle layer in cerebral arteries. Like most signaling pathways, the Notch signaling axis is influenced by complex interactions with mediators of other signaling pathways. This complexity is also compounded by different members of the Notch family having both overlapping and unique functions. Thus, it is vital to fully understand the roles and interactions of each Notch family member in order to effectively and specifically target their exact contributions to vascular disease. In this chapter, we will review the Notch signaling pathway in vascular smooth muscle cells as it relates to vascular development and human disease.
Collapse
Affiliation(s)
- J T Baeten
- The Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, United States
| | - B Lilly
- The Center for Cardiovascular Research and The Heart Center at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
27
|
Preuße K, Tveriakhina L, Schuster-Gossler K, Gaspar C, Rosa AI, Henrique D, Gossler A, Stauber M. Context-Dependent Functional Divergence of the Notch Ligands DLL1 and DLL4 In Vivo. PLoS Genet 2015; 11:e1005328. [PMID: 26114479 PMCID: PMC4482573 DOI: 10.1371/journal.pgen.1005328] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 06/01/2015] [Indexed: 11/18/2022] Open
Abstract
Notch signalling is a fundamental pathway that shapes the developing embryo and sustains adult tissues by direct communication between ligand and receptor molecules on adjacent cells. Among the ligands are two Delta paralogues, DLL1 and DLL4, that are conserved in mammals and share a similar structure and sequence. They activate the Notch receptor partly in overlapping expression domains where they fulfil redundant functions in some processes (e.g. maintenance of the crypt cell progenitor pool). In other processes, however, they appear to act differently (e.g. maintenance of foetal arterial identity) raising the questions of how similar DLL1 and DLL4 really are and which mechanism causes the apparent context-dependent divergence. By analysing mice that conditionally overexpress DLL1 or DLL4 from the same genomic locus (Hprt) and mice that express DLL4 instead of DLL1 from the endogenous Dll1 locus (Dll1Dll4ki), we found functional differences that are tissue-specific: while DLL1 and DLL4 act redundantly during the maintenance of retinal progenitors, their function varies in the presomitic mesoderm (PSM) where somites form in a Notch-dependent process. In the anterior PSM, every cell expresses both Notch receptors and ligands, and DLL1 is the only activator of Notch while DLL4 is not endogenously expressed. Transgenic DLL4 cannot replace DLL1 during somitogenesis and in heterozygous Dll1Dll4ki/+ mice, the Dll1Dll4ki allele causes a dominant segmentation phenotype. Testing several aspects of the complex Notch signalling system in vitro, we found that both ligands have a similar trans-activation potential but that only DLL4 is an efficient cis-inhibitor of Notch signalling, causing a reduced net activation of Notch. These differential cis-inhibitory properties are likely to contribute to the functional divergence of DLL1 and DLL4.
Collapse
Affiliation(s)
- Kristina Preuße
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Hannover, Germany
| | - Lena Tveriakhina
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Hannover, Germany
| | - Karin Schuster-Gossler
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Hannover, Germany
| | - Cláudia Gaspar
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Alexandra Isabel Rosa
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Domingos Henrique
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Achim Gossler
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Hannover, Germany
| | - Michael Stauber
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Hannover, Germany
| |
Collapse
|
28
|
Applebaum M, Kalcheim C. Mechanisms of myogenic specification and patterning. Results Probl Cell Differ 2015; 56:77-98. [PMID: 25344667 DOI: 10.1007/978-3-662-44608-9_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesodermal somites are initially composed of columnar cells arranged as a pseudostratified epithelium that undergoes sequential and spatially restricted changes to generate the sclerotome and dermomyotome, intermediate structures that develop into vertebrae, striated muscles of the body and limbs, dermis, smooth muscle, and endothelial cells. Regional cues were elucidated that impart differential traits upon the originally multipotent progenitors. How do somite cells and their intermediate progenitors interpret these extrinsic cues and translate them into various levels and/or modalities of intracellular signaling that lead to differential gene expression profiles remains a significant challenge. So is the understanding of how differential fate specification relates to complex cellular migrations prefiguring the formation of body muscles and vertebrae. Research in the past years has largely transited from a descriptive phase in which the lineages of distinct somite-derived progenitors and their cellular movements were traced to a more mechanistic understanding of the local function of genes and regulatory networks underlying lineage segregation and tissue organization. In this chapter, we focus on some major advances addressing the segregation of lineages from the dermomyotome, while discussing both cellular as well as molecular mechanisms, where possible.
Collapse
Affiliation(s)
- Mordechai Applebaum
- Department of Medical Neurobiology, IMRIC and ELSC-Hebrew University-Hadassah Medical School, Jerusalem, 9101201, 12272, Israel,
| | | |
Collapse
|
29
|
Notch signaling regulates myogenic regenerative capacity of murine and human mesoangioblasts. Cell Death Dis 2014; 5:e1448. [PMID: 25299773 PMCID: PMC4237240 DOI: 10.1038/cddis.2014.401] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 07/28/2014] [Accepted: 08/26/2014] [Indexed: 01/12/2023]
Abstract
Somatic stem cells hold attractive potential for the treatment of muscular dystrophies (MDs). Mesoangioblasts (MABs) constitute a myogenic subset of muscle pericytes and have been shown to efficiently regenerate dystrophic muscles in mice and dogs. In addition, HLA-matched MABs are currently being tested in a phase 1 clinical study on Duchenne MD patients (EudraCT #2011-000176-33). Many reports indicate that the Notch pathway regulates muscle regeneration and satellite cell commitment. However, little is known about Notch-mediated effects on other resident myogenic cells. To possibly potentiate MAB-driven regeneration in vivo, we asked whether Notch signaling played a pivotal role in regulating MAB myogenic capacity. Through different approaches of loss- and gain-of-function in murine and human MABs, we determined that the interplay between Delta-like ligand 1 (Dll1)-activated Notch1 and Mef2C supports MAB commitment in vitro and ameliorates engraftment and functional outcome after intra-arterial delivery in dystrophic mice. Furthermore, using a transgenic mouse model of conditional Dll1 deletion, we demonstrated that Dll1 ablation, either on the injected cells, or on the receiving muscle fibers, impairs MAB regenerative potential. Our data corroborate the perspective of advanced combinations of cell therapy and signaling tuning to enhance therapeutic efficaciousness of somatic stem cells.
Collapse
|
30
|
Zhang P, Yan X, Chen Y, Yang Z, Han H. Notch signaling in blood vessels: from morphogenesis to homeostasis. SCIENCE CHINA-LIFE SCIENCES 2014; 57:774-80. [PMID: 25104449 DOI: 10.1007/s11427-014-4716-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 06/16/2013] [Indexed: 12/28/2022]
Abstract
Notch signaling is an evolutionarily conserved intercellular signaling pathway that plays numerous crucial roles in vascular development and physiology. Compelling evidence indicates that Notch signaling is vital for vascular morphogenesis including arterial and venous differentiation and endothelial tip and stalk cell specification during sprouting angiogenesis and also vessel maturation featured by mural cell differentiation and recruitment. Notch signaling is also required for vascular homeostasis in adults by keeping quiescent phalanx cells from re-entering cell cycle and by modulating the behavior of endothelial progenitor cells. We will summarize recent advances of Notch pathway in vascular biology with special emphasis on the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | | | |
Collapse
|
31
|
Forbes SJ, Rosenthal N. Preparing the ground for tissue regeneration: from mechanism to therapy. Nat Med 2014; 20:857-69. [PMID: 25100531 DOI: 10.1038/nm.3653] [Citation(s) in RCA: 413] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/14/2014] [Indexed: 12/12/2022]
Abstract
Chronic diseases confer tissue and organ damage that reduce quality of life and are largely refractory to therapy. Although stem cells hold promise for treating degenerative diseases by 'seeding' injured tissues, the regenerative capacity of stem cells is influenced by regulatory networks orchestrated by local immune responses to tissue damage, with macrophages being a central component of the injury response and coordinator of tissue repair. Recent research has turned to how cellular and signaling components of the local stromal microenvironment (the 'soil' to the stem cells' seed), such as local inflammatory reactions, contribute to successful tissue regeneration. This Review discusses the basic principles of tissue regeneration and the central role locally acting components may play in the process. Application of seed-and-soil concepts to regenerative medicine strengthens prospects for developing cell-based therapies or for promotion of endogenous repair.
Collapse
Affiliation(s)
- Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Nadia Rosenthal
- 1] National Heart and Lung Institute, Imperial College London, London, UK. [2] Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Applebaum M, Ben-Yair R, Kalcheim C. Segregation of striated and smooth muscle lineages by a Notch-dependent regulatory network. BMC Biol 2014; 12:53. [PMID: 25015411 PMCID: PMC4260679 DOI: 10.1186/s12915-014-0053-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Indexed: 12/31/2022] Open
Abstract
Background Lineage segregation from multipotent epithelia is a central theme in development and in adult stem cell plasticity. Previously, we demonstrated that striated and smooth muscle cells share a common progenitor within their epithelium of origin, the lateral domain of the somite-derived dermomyotome. However, what controls the segregation of these muscle subtypes remains unknown. We use this in vivo bifurcation of fates as an experimental model to uncover the underlying mechanisms of lineage diversification from bipotent progenitors. Results Using the strength of spatio-temporally controlled gene missexpression in avian embryos, we report that Notch harbors distinct pro-smooth muscle activities depending on the duration of the signal; short periods prevent striated muscle development and extended periods, through Snail1, promote cell emigration from the dermomyotome towards a smooth muscle fate. Furthermore, we define a Muscle Regulatory Network, consisting of Id2, Id3, FoxC2 and Snail1, which acts in concert to promote smooth muscle by antagonizing the pro-myogenic activities of Myf5 and Pax7, which induce striated muscle fate. Notch and BMP closely regulate the network and reciprocally reinforce each other’s signal. In turn, components of the network strengthen Notch signaling, while Pax7 silences this signaling. These feedbacks augment the robustness and flexibility of the network regulating muscle subtype segregation. Conclusions Our results demarcate the details of the Muscle Regulatory Network, underlying the segregation of muscle sublineages from the lateral dermomyotome, and exhibit how factors within the network promote the smooth muscle at the expense of the striated muscle fate. This network acts as an exemplar demonstrating how lineage segregation occurs within epithelial primordia by integrating inputs from competing factors.
Collapse
|
33
|
Optimization of stem cell therapy for clinical trials of degenerative skin and muscle diseases (OptiStem). HUM GENE THER CL DEV 2014; 25:68-70. [PMID: 24933567 DOI: 10.1089/humc.2014.2510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
34
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Olson JD, Mintz A, Delbono O. Type-2 pericytes participate in normal and tumoral angiogenesis. Am J Physiol Cell Physiol 2014; 307:C25-38. [PMID: 24788248 DOI: 10.1152/ajpcell.00084.2014] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue growth and function depend on vascularization, and vascular insufficiency or excess exacerbates many human diseases. Identification of the biological processes involved in angiogenesis will dictate strategies to modulate reduced or excessive vessel formation. We examine the essential role of pericytes. Their heterogeneous morphology, distribution, origins, and physiology have been described. Using double-transgenic Nestin-GFP/NG2-DsRed mice, we identified two pericyte subsets. We found that Nestin-GFP(-)/NG2-DsRed(+) (type-1) and Nestin-GFP(+)/NG2-DsRed(+) (type-2) pericytes attach to the walls of small and large blood vessels in vivo; in vitro, type-2, but not type-1, pericytes spark endothelial cells to form new vessels. Matrigel assay showed that only type-2 pericytes participate in normal angiogenesis. Moreover, when cancer cells were transplanted into Nestin-GFP/NG2-DsRed mice, type-1 pericytes did not penetrate the tumor, while type-2 pericytes were recruited during its angiogenesis. As inhibition of angiogenesis is a promising strategy in cancer therapy, type-2 pericytes may provide a cellular target susceptible to signaling and pharmacological manipulation in treating malignancy. This work also reports the potential of type-2 pericytes to improve blood perfusion in ischemic hindlimbs, indicating their potential for treating ischemic illnesses.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John D Olson
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina; Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina;
| |
Collapse
|
35
|
|
36
|
Fukada SI, Ma Y, Ohtani T, Watanabe Y, Murakami S, Yamaguchi M. Isolation, characterization, and molecular regulation of muscle stem cells. Front Physiol 2013; 4:317. [PMID: 24273513 PMCID: PMC3824104 DOI: 10.3389/fphys.2013.00317] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/14/2013] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle has great regenerative capacity which is dependent on muscle stem cells, also known as satellite cells. A loss of satellite cells and/or their function impairs skeletal muscle regeneration and leads to a loss of skeletal muscle power; therefore, the molecular mechanisms for maintaining satellite cells in a quiescent and undifferentiated state are of great interest in skeletal muscle biology. Many studies have demonstrated proteins expressed by satellite cells, including Pax7, M-cadherin, Cxcr4, syndecan3/4, and c-met. To further characterize satellite cells, we established a method to directly isolate satellite cells using a monoclonal antibody, SM/C-2.6. Using SM/C-2.6 and microarrays, we measured the genes expressed in quiescent satellite cells and demonstrated that Hesr3 may complement Hesr1 in generating quiescent satellite cells. Although Hesr1- or Hesr3-single knockout mice show a normal skeletal muscle phenotype, including satellite cells, Hesr1/Hesr3-double knockout mice show a gradual decrease in the number of satellite cells and increase in regenerative defects dependent on satellite cell numbers. We also observed that a mouse's genetic background affects the regenerative capacity of its skeletal muscle and have established a line of DBA/2-background mdx mice that has a much more severe phenotype than the frequently used C57BL/10-mdx mice. The phenotype of DBA/2-mdx mice also seems to depend on the function of satellite cells. In this review, we summarize the methodology of direct isolation, characterization, and molecular regulation of satellite cells based on our results. The relationship between the regenerative capacity of satellite cells and progression of muscular disorders is also summarized. In the last part, we discuss application of the accumulating scientific information on satellite cells to treatment of patients with muscular disorders.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University Osaka, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Increasing attention is currently devoted to the multiple roles that pericytes (also defined as mural, Rouget, or perivascular cells) may play during angiogenesis, vascular homeostasis, and pathology. Many recent excellent reviews thoroughly address these topics (see below); hence, we will not discuss them in detail here. However, not much is known about origin, heterogeneity, gene expression, and developmental potential of pericytes during fetal and postnatal development. This is likely because of the paucity of markers expressed by pericytes and the absence of truly unique ones. Thus, in vivo identification and ex perspective isolation are challenging and explain the relative little data available in comparison with neighbor but far more characterized cells such as the endothelium. Despite this preliminary knowledge, we will propose that contribution to growing mesoderm tissues may be an important role for pericytes. Thus, their ability to contribute to tissue regeneration may be a consequence of their role in tissue growth. However, in a severely damaged or diseased tissue, acute or chronic inflammation likely results in the production of signaling molecules that are different from those present in developing tissues, thus explaining why pericytes are easily diverted from a regenerative to a fibrotic fate.
Collapse
Affiliation(s)
- Ornella Cappellari
- Department of Cell and Developmental Biology, University College London, United Kingdom
| | | |
Collapse
|
38
|
Montarras D, L'honoré A, Buckingham M. Lying low but ready for action: the quiescent muscle satellite cell. FEBS J 2013; 280:4036-50. [DOI: 10.1111/febs.12372] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/24/2013] [Accepted: 05/28/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Didier Montarras
- Department of Developmental and Stem Cell Biology; CNRS URA 2578; Institut Pasteur; Paris; France
| | - Aurore L'honoré
- Department of Developmental and Stem Cell Biology; CNRS URA 2578; Institut Pasteur; Paris; France
| | - Margaret Buckingham
- Department of Developmental and Stem Cell Biology; CNRS URA 2578; Institut Pasteur; Paris; France
| |
Collapse
|
39
|
Abstract
Pericytes, typically attached to the walls of microvessels in almost all organs, interact with endothelial cells and take part in diverse biological processes, e.g. blood vessel regulation and tissue repair. This suggests that pericytes harbor a remarkable degree of cellular plasticity, which could potentially be employed for the treatment of diseases affecting diverse tissues such as the skeletal muscle and the central nervous system. Here, we follow pericytes on their journey across Waddington's epigenetic landscape, descending from their origin, along a path guided by environmental signals or ectopic transcription factors, at the end of which they acquire a new identity, e.g. muscle or nerve cells. The central theme of this review is the question of whether pericytes can be enticed to differentiate into whatever cell type is needed, and thus provide an endogenous cellular source for treating as yet incurable diseases--like a magic bullet.
Collapse
Affiliation(s)
- Marisa Karow
- Department of Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich, Munich, Germany.
| |
Collapse
|
40
|
Abstract
Microvascular pericytes are able to generate multiple mesenchymal cell types, including skeletal muscle myoblasts. Cappellari et al. (2013) report in this issue of Developmental Cell that myoblasts can return the favor by generating pericytes via the action of Dll4 Notch ligand and PDGF-BB.
Collapse
Affiliation(s)
- William B Stallcup
- Tumor Microenvironment Program, Cancer Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
41
|
Biressi S, Bjornson CRR, Carlig PMM, Nishijo K, Keller C, Rando TA. Myf5 expression during fetal myogenesis defines the developmental progenitors of adult satellite cells. Dev Biol 2013; 379:195-207. [PMID: 23639729 DOI: 10.1016/j.ydbio.2013.04.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 03/21/2013] [Accepted: 04/09/2013] [Indexed: 01/06/2023]
Abstract
Myf5 is a member of the muscle-specific determination genes and plays a critical role in skeletal muscle development. Whereas the expression of Myf5 during embryonic and fetal myogenesis has been extensively studied, its expression in progenitors that will ultimately give rise to adult satellite cells, the stem cells responsible for muscle repair, is still largely unexplored. To investigate this aspect, we have generated a mouse strain carrying a CreER coding sequence in the Myf5 locus. In this strain, Tamoxifen-inducible Cre activity parallels endogenous Myf5 expression. Combining Myf5(CreER) and Cre reporter alleles, we were able to evaluate the contribution of cells expressing Myf5 at distinct developmental stages to the pool of satellite cells in adult hindlimb muscles. Although it was possible to trace back the origin of some rare satellite cells to a subpopulation of Myf5(+ve) progenitors in the limb buds at the late embryonic stage (∼E12), a significant number of satellite cells arise from cells which expressed Myf5 for the first time at the fetal stage (∼E15). These studies provide direct evidence that adult satellite cells derive from progenitors that first express the myogenic determination gene Myf5 during fetal stages of myogenesis.
Collapse
Affiliation(s)
- Stefano Biressi
- Paul F. Glenn Laboratories for the Biology of Aging and Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|