1
|
Wu Y, Song Y, Soto J, Hoffman T, Lin X, Zhang A, Chen S, Massad RN, Han X, Qi D, Yeh KW, Fang Z, Eoh J, Gu L, Rowat AC, Gu Z, Li S. Viscoelastic Extracellular Matrix Enhances Epigenetic Remodeling and Cellular Plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.14.589442. [PMID: 38659850 PMCID: PMC11042188 DOI: 10.1101/2024.04.14.589442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Extracellular matrices of living tissues exhibit viscoelastic properties, yet how these properties regulate chromatin and the epigenome remains unclear. Here, we show that viscoelastic substrates induce changes in nuclear architecture and epigenome, with more pronounced effects on softer surfaces. Fibroblasts on viscoelastic substrates display larger nuclei, lower chromatin compaction, and differential expression of distinct sets of genes related to the cytoskeleton and nuclear function compared to those on purely elastic surfaces. Slow-relaxing viscoelastic substrates reduce lamin A/C expression and enhance nuclear remodeling. These structural changes are accompanied by a global increase in euchromatin marks and local increase in chromatin accessibility at cis-regulatory elements associated with neuronal and pluripotent genes. Consequently, viscoelastic substrates improve the reprogramming efficiency from fibroblasts into neurons and induced pluripotent stem cells. Collectively, our findings unravel the roles of matrix viscoelasticity in epigenetic regulation and cell reprogramming, with implications for designing smart materials for cell fate engineering.
Collapse
|
2
|
Sharma A, Steger RF, Li JM, Baude JA, Heom KA, Dey SS, Stowers RS. Sp1 mechanotransduction regulates breast cancer cell invasion in response to multiple tumor-mimicking extracellular matrix cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643983. [PMID: 40166320 PMCID: PMC11957027 DOI: 10.1101/2025.03.18.643983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Breast cancer progression is marked by extracellular matrix (ECM) remodeling, including increased stiffness, faster stress relaxation, and elevated collagen levels. In vitro experiments have revealed a role for each of these factors to individually promote malignant behavior, but their combined effects remain unclear. To address this, we developed alginate-collagen hydrogels with independently tunable stiffness, stress relaxation, and collagen density. We show that these combined tumor-mimicking ECM cues reinforced invasive morphologies and promoted spheroid invasion in breast cancer and mammary epithelial cells. High stiffness and low collagen density in slow-relaxing matrices led to the greatest cell migration speed and displacement. RNA-seq revealed Sp1 target gene enrichment in response to both individual and combined ECM cues, with a greater enrichment observed under multiple cues. Notably, high expression of Sp1 target genes upregulated by fast stress relaxation correlated with poor patient survival. Mechanistically, we found that phosphorylated-Sp1 (T453) was increasingly located in the nucleus in stiff and/or fast relaxing matrices, which was regulated by PI3K and ERK1/2 signaling, as well as actomyosin contractility. This study emphasizes how multiple ECM cues in complex microenvironments reinforce malignant traits and supports an emerging role for Sp1 as a mechanoresponsive transcription factor.
Collapse
Affiliation(s)
- Abhishek Sharma
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Rowan F Steger
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jen M Li
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jane A Baude
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Kellie A Heom
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Siddharth S Dey
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
3
|
Sharma V, Adebowale K, Gong Z, Chaudhuri O, Shenoy VB. Glassy Adhesion Dynamics Govern Transitions Between Sub-Diffusive and Super-Diffusive Cell Migration on Viscoelastic Substrates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642113. [PMID: 40161659 PMCID: PMC11952418 DOI: 10.1101/2025.03.11.642113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cell migration is pivotal in cancer metastasis, where cells navigate the extracellular matrix (ECM) and invade distant tissues. While the ECM is viscoelastic-exhibiting time-dependent stress relaxation-its influence on cell migration remains poorly understood. Here, we employ an integrated experimental and modeling approach to investigate filopodial cancer cell migration on viscoelastic substrates and uncover a striking transition from sub-diffusive to super-diffusive behavior driven by the substrate's viscous relaxation timescale. Conventional motor-clutch based migration models fail to capture these anomalous migration modes, as they overlook the complex adhesion dynamics shaped by broad distribution of adhesion lifetimes. To address this, we develop a glassy motor-clutch model that incorporates the rugged energy landscape of adhesion clusters, where multiple metastable states yield long-tailed adhesion timescales. Our model reveals that migration dynamics are governed by the interplay between cellular and substrate timescales: slow-relaxing substrates prolong trapping, leading to sub-diffusion, while fast-relaxing substrates promote larger steps limiting trapping, leading to super-diffusion. Additionally, we uncover the role of actin polymerization and contractility in modulating adhesion dynamics and driving anomalous migration. These findings establish a mechanistic framework linking substrate viscoelasticity to cell motility, with implications for metastasis and cancer progression.
Collapse
|
4
|
Ren Q, Han X, Gong S, Zhang S, Ba T, Zhao Y, Li Y, Wang Y, Zhou X, Li Y, Ji L. Clinical features and search for genetic determinants of postprandial hypoglycaemia. Endocr Connect 2025; 14:e240409. [PMID: 39495171 PMCID: PMC11728931 DOI: 10.1530/ec-24-0409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/07/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Objective To test whether postprandial hypoglycaemia is an extreme and repeatable phenotype of glucose metabolism. We also explored the genetic determinants of this phenotype. Design and methods We conducted this study using data from the Pinggu Metabolic Disease Study database (n = 3,345). We selected subjects after an oral glucose tolerance test (OGTT) (2 h glucose < 3 mmol/L) and compared their clinical features with those of subjects with normal glucose tolerance (NGT). In addition, we selected 75 subjects as a super-healthy control group. Whole-exome sequencing (WES) was performed on subjects with postprandial hypoglycaemic and super-healthy controls. We also evaluated several candidate genes believed to be important in pancreatic hypoglycaemia. Results We found 13 participants (0.39%) who had an OGTT (2 h glucose < 3 mmol/L). Ten of these patients were men (76.9%). All 13 participants had insulin >3 μU/mL when postprandial blood glucose levels were <3 mmol/L. WES analysis identified one gene, paternally expressed 3 (PEG3), which had three rare mutations in four patients (30.8%). Minor allele frequencies of rare PEG3 mutations were significantly higher in subjects with postprandial hypoglycaemia than in super-healthy controls. Among the four subjects with PEG3 gene mutations, 71.4% were men, and their body mass index was significantly lower than that of the NGT group. Conclusions Postprandial hypoglycaemia is an extreme and reproducible phenotype in the general population. PEG3 mutations may represent a potential genetic aetiology for postprandial hypoglycaemia. Further research with larger and more diverse populations and a broader genetic focus is needed to understand the genetic basis of postprandial hypoglycaemia.
Collapse
Affiliation(s)
- Qian Ren
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | - Xueyao Han
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | - Siqian Gong
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | - Simin Zhang
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | | | | | - Yating Li
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | - Yan’ai Wang
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | - Xianghai Zhou
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| | - Yufeng Li
- Department of Endocrinology, Beijing Pinggu Hospital, Beijing, China
| | - Linong Ji
- Department of Endocrinology, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
5
|
Charbonier F, Zhu J, Slyman R, Allan C, Chaudhuri O. Substrate stress relaxation regulates monolayer fluidity and leader cell formation for collectively migrating epithelia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609529. [PMID: 39253481 PMCID: PMC11383040 DOI: 10.1101/2024.08.26.609529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Collective migration of epithelial tissues is a critical feature of developmental morphogenesis and tissue homeostasis. Coherent motion of cell collectives requires large scale coordination of motion and force generation and is influenced by mechanical properties of the underlying substrate. While tissue viscoelasticity is a ubiquitous feature of biological tissues, its role in mediating collective cell migration is unclear. Here, we have investigated the impact of substrate stress relaxation on the migration of micropatterned epithelial monolayers. Epithelial monolayers exhibit faster collective migration on viscoelastic alginate substrates with slower relaxation timescales, which are more elastic, relative to substrates with faster stress relaxation, which exhibit more viscous loss. Faster migration on slow-relaxing substrates is associated with reduced substrate deformation, greater monolayer fluidity, and enhanced leader cell formation. In contrast, monolayers on fast-relaxing substrates generate substantial substrate deformations and are more jammed within the bulk, with reduced formation of transient lamellipodial protrusions past the monolayer edge leading to slower overall expansion. This work reveals features of collective epithelial dynamics on soft, viscoelastic materials and adds to our understanding of cell-substrate interactions at the tissue scale. Significance Statement Groups of cells must coordinate their movements in order to sculpt organs during development and maintain tissues. The mechanical properties of the underlying substrate on which cells reside are known to influence key aspects of single and collective cell migration. Despite being a nearly universal feature of biological tissues, the role of viscoelasticity (i.e., fluid-like and solid-like behavior) in collective cell migration is unclear. Using tunable engineered biomaterials, we demonstrate that sheets of epithelial cells display enhanced migration on slower-relaxing (more elastic) substrates relative to faster-relaxing (more viscous) substrates. Building our understanding of tissue-substrate interactions and collective cell dynamics provides insights into approaches for tissue engineering and regenerative medicine, and therapeutic interventions to promote health and treat disease.
Collapse
|
6
|
Narasimhan BN, Fraley SI. Degradability tunes ECM stress relaxation and cellular mechanics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605514. [PMID: 39131364 PMCID: PMC11312499 DOI: 10.1101/2024.07.28.605514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
In native extracellular matrices (ECM), cells can use matrix metalloproteinases (MMPs) to degrade and remodel their surroundings. Likewise, synthetic matrices have been engineered to facilitate MMP-mediated cleavage that enables cell spreading, migration, and interactions. However, the intersection of matrix degradability and mechanical properties has not been fully considered. We hypothesized that immediate mechanical changes result from the action of MMPs on the ECM and that these changes are sensed by cells. Using atomic force microscopy (AFM) to measure cell-scale mechanical properties, we find that both fibrillar collagen and synthetic degradable matrices exhibit enhanced stress relaxation after MMP exposure. Cells respond to these relaxation differences by altering their spreading and focal adhesions. We demonstrate that stress relaxation can be tuned through the rational design of matrix degradability. These findings establish a fundamental link between matrix degradability and stress relaxation, which may impact a range of biological applications.
Collapse
Affiliation(s)
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
7
|
Akat A, Karaöz E. Cell Therapy Strategies on Duchenne Muscular Dystrophy: A Systematic Review of Clinical Applications. Stem Cell Rev Rep 2024; 20:138-158. [PMID: 37955832 DOI: 10.1007/s12015-023-10653-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is an inherited genetic disorder characterized by progressive degeneration of muscle tissue, leading to functional disability and premature death. Despite extensive research efforts, the discovery of a cure for DMD continues to be elusive, emphasizing the need to investigate novel treatment approaches. Cellular therapies have emerged as prospective approaches to address the underlying pathophysiology of DMD. This review provides an examination of the present situation regarding cell-based therapies, including CD133 + cells, muscle precursor cells, mesoangioblasts, bone marrow-derived mononuclear cells, mesenchymal stem cells, cardiosphere-derived cells, and dystrophin-expressing chimeric cells. A total of 12 studies were found eligible to be included as they were completed cell therapy clinical trials, clinical applications, or case reports with quantitative results. The evaluation encompassed an examination of limitations and potential advancements in this particular area of research, along with an assessment of the safety and effectiveness of cell-based therapies in the context of DMD. In general, the available data indicates that diverse cell therapy approaches may present a new, safe, and efficacious treatment modality for patients diagnosed with DMD. However, further studies are required to comprehensively understand the most advantageous treatment approach and therapeutic capacity.
Collapse
Affiliation(s)
- Ayberk Akat
- Life Park Hospital, Cellular and Biological Products Manufacturing Center, Ragıp Kenan Sok. No:8, Ortakoy, 99010, Nicosia (Lefkosa), Cyprus.
| | - Erdal Karaöz
- Liv Hospital Ulus, Regenerative Medicine and Stem Cell Center, Istanbul, Turkey
| |
Collapse
|
8
|
Li H, Kuhn M, Kelly RA, Singh A, Palanivel KK, Salama I, De Ieso ML, Stamer WD, Ganapathy PS, Herberg S. Targeting YAP mechanosignaling to ameliorate stiffness-induced Schlemm's canal cell pathobiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556840. [PMID: 37781615 PMCID: PMC10541092 DOI: 10.1101/2023.09.08.556840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Pathologic alterations in the biomechanical properties of the Schlemm's canal (SC) inner wall endothelium and its immediate vicinity are strongly associated with ocular hypertension in glaucoma due to decreased outflow facility. Specifically, the underlying trabecular meshwork is substantially stiffer in glaucomatous eyes compared to that from normal eyes. This raises the possibility of a critical involvement of mechanotransduction processes in driving SC cell dysfunction. Yes-associated protein (YAP) has emerged as a key contributor to glaucoma pathogenesis. However, the molecular underpinnings of SC cell YAP mechanosignaling in response to glaucomatous extracellular matrix (ECM) stiffening are not well understood. Using a novel biopolymer hydrogel that facilitates dynamic and reversible stiffness tuning, we investigated how ECM stiffening modulates YAP activity in primary human SC cells, and whether disruption of YAP mechanosignaling attenuates SC cell pathobiology and increases ex vivo outflow facility. We demonstrated that ECM stiffening drives pathologic YAP activation and cytoskeletal reorganization in SC cells, which was fully reversible by matrix softening in a distinct time-dependent manner. Furthermore, we showed that pharmacologic or genetic disruption of YAP mechanosignaling abrogates stiffness-induced SC cell dysfunction involving altered cytoskeletal and ECM remodeling. Lastly, we found that perfusion of the clinically-used, small molecule YAP inhibitor verteporfin (without light activation) increases ex vivo outflow facility in normal mouse eyes. Collectively, our data provide new evidence for a pathologic role of aberrant YAP mechanosignaling in SC cell dysfunction and suggest that YAP inhibition has therapeutic value for treating ocular hypertension in glaucoma.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Megan Kuhn
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
| | - Ruth A. Kelly
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
| | - Ayushi Singh
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Kavipriya Kovai Palanivel
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Izzy Salama
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael L. De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Preethi S. Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA
| |
Collapse
|
9
|
Giarratana N, Conti F, Rinvenuti L, Ronzoni F, Sampaolesi M. State of the Art Procedures for the Isolation and Characterization of Mesoangioblasts. Methods Mol Biol 2023; 2640:99-115. [PMID: 36995590 DOI: 10.1007/978-1-0716-3036-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Adult skeletal muscle is a dynamic tissue able to regenerate quite efficiently, thanks to the presence of stem cell machinery. Besides the quiescent satellite cells that are activated upon injury or paracrine factors, other stem cells are described to be directly or indirectly involved in adult myogenesis. Mesoangioblasts (MABs) are vessel-associated stem cells originally isolated from embryonic dorsal aorta and, at later stages, from the adult muscle interstitium expressing pericyte markers. Adult MABs entered clinical trials for the treatment of Duchenne muscular dystrophy and the transcriptome of human fetal MABs has been described. In addition, single cell RNA-seq analyses provide novel information on adult murine MABs and more in general in interstitial muscle stem cells. This chapter provides state-of-the-art techniques to isolate and characterize murine MABs, fetal and adult human MABs.
Collapse
Affiliation(s)
- Nefele Giarratana
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Filippo Conti
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Lorenza Rinvenuti
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Flavio Ronzoni
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology Unit, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - 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 of Rome, Rome, Italy.
| |
Collapse
|
10
|
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
|
11
|
Tanaka K, Besson V, Rivagorda M, Oury F, Marazzi G, Sassoon DA. Paternally expressed gene 3 (Pw1/Peg3) promotes sexual dimorphism in metabolism and behavior. PLoS Genet 2022; 18:e1010003. [PMID: 35025875 PMCID: PMC8791484 DOI: 10.1371/journal.pgen.1010003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 01/26/2022] [Accepted: 12/20/2021] [Indexed: 01/06/2023] Open
Abstract
The paternally expressed gene 3 (Pw1/Peg3) is a mammalian-specific parentally imprinted gene expressed in stem/progenitor cells of the brain and endocrine tissues. Here, we compared phenotypic characteristics in Pw1/Peg3 deficient male and female mice. Our findings indicate that Pw1/Peg3 is a key player for the determination of sexual dimorphism in metabolism and behavior. Mice carrying a paternally inherited Pw1/Peg3 mutant allele manifested postnatal deficits in GH/IGF dependent growth before weaning, sex steroid dependent masculinization during puberty, and insulin dependent fat accumulation in adulthood. As a result, Pw1/Peg3 deficient mice develop a sex-dependent global shift of body metabolism towards accelerated adiposity, diabetic-like insulin resistance, and fatty liver. Furthermore, Pw1/Peg3 deficient males displayed reduced social dominance and competitiveness concomitant with alterations in the vasopressinergic architecture in the brain. This study demonstrates that Pw1/Peg3 provides an epigenetic context that promotes male-specific characteristics through sex steroid pathways during postnatal development. Pw1/Peg3 is under parental specific epigenetic regulation. We propose that Pw1/Peg3 confers a selective advantage in mammals by regulating sexual dimorphism. To address this question, we examined the consequences of Pw1/Peg3 loss of function in mice in an age- and sex-dependent context and found that Pw1/Peg3 mutants display reduced sexual dimorphism in growth, metabolism and behaviors. Our findings support the intralocus sexual conflict model of genomic imprinting where it contributes in sexual differentiation. Furthermore, our observations provide a unifying role of sex steroid signaling as a common property of Pw1/Peg3 expressing stem/progenitor cells and differentiated endocrine cells, both of which remain proliferative in response to gonadal hormones in adult life.
Collapse
Affiliation(s)
- Karo Tanaka
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Vanessa Besson
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Manon Rivagorda
- Hormonal Regulation of Brain Development and Functions, INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Franck Oury
- Hormonal Regulation of Brain Development and Functions, INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
| | - David A. Sassoon
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), INSERM U1166, University of Pierre and Marie Curie Paris VI, Paris, France
- * E-mail:
| |
Collapse
|
12
|
Ritso M, Tung LW, Rossi FMV. Emerging skeletal muscle stromal cell diversity: Functional divergence in fibro/adipogenic progenitor and mural cell populations. Exp Cell Res 2022; 410:112947. [PMID: 34822813 DOI: 10.1016/j.yexcr.2021.112947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 12/27/2022]
Abstract
While the majority of healthy skeletal muscle consists of multinucleated syncytial repetitive contractile myofibers, repaired by skeletal muscle stem cells when damaged, the maintenance of muscle function also requires a range of tissue-resident stromal populations. In fact, the careful orchestration of damage response processes upon muscle injury relies heavily on stromal cell contribution for effective repair. The two main types of muscle-resident stromal cells are fibro/adipogenic progenitors and mural cells. The latter is comprised of pericytes and vascular smooth muscle cells. Recent publications identifying common markers for stromal cell populations have allowed investigating population dynamics throughout the regenerative process at a higher resolution. Mounting evidence now suggests that subpopulations with distinct roles may exist among stromal cells. In various degenerative muscle wasting conditions, critical cross-talk and spatial signalling amongst various cell populations become dysregulated. This can result in the failure to curb pathological fibro/adipogenic progenitor proliferation and propensity for laying down excessive extracellular matrix, which in turn leads to fibrotic infiltration, reduced contractile units and gradual decline in muscle function. Restoration of physiologically appropriate stromal cell function is therefore just as crucial for therapeutic targeting as the homeostatic maintenance of muscle function.
Collapse
Affiliation(s)
- Morten Ritso
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
13
|
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
|
14
|
Contessotto P, Orbanić D, Da Costa M, Jin C, Owens P, Chantepie S, Chinello C, Newell J, Magni F, Papy-Garcia D, Karlsson NG, Kilcoyne M, Dockery P, Rodríguez-Cabello JC, Pandit A. Elastin-like recombinamers-based hydrogel modulates post-ischemic remodeling in a non-transmural myocardial infarction in sheep. Sci Transl Med 2021; 13:13/581/eaaz5380. [PMID: 33597263 DOI: 10.1126/scitranslmed.aaz5380] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 09/30/2020] [Accepted: 01/27/2021] [Indexed: 01/11/2023]
Abstract
Ischemic heart disease is a leading cause of mortality due to irreversible damage to cardiac muscle. Inspired by the post-ischemic microenvironment, we devised an extracellular matrix (ECM)-mimicking hydrogel using catalyst-free click chemistry covalent bonding between two elastin-like recombinamers (ELRs). The resulting customized hydrogel included functional domains for cell adhesion and protease cleavage sites, sensitive to cleavage by matrix metalloproteases overexpressed after myocardial infarction (MI). The scaffold permitted stromal cell invasion and endothelial cell sprouting in vitro. The incidence of non-transmural infarcts has increased clinically over the past decade, and there is currently no treatment preventing further functional deterioration in the infarcted areas. Here, we have developed a clinically relevant ovine model of non-transmural infarcts induced by multiple suture ligations. Intramyocardial injections of the degradable ELRs-hydrogel led to complete functional recovery of ejection fraction 21 days after the intervention. We observed less fibrosis and more angiogenesis in the ELRs-hydrogel-treated ischemic core region compared to the untreated animals, as validated by the expression, proteomic, glycomic, and histological analyses. These findings were accompanied by enhanced preservation of GATA4+ cardiomyocytes in the border zone of the infarct. We propose that our customized ECM favors cardiomyocyte preservation in the border zone by modulating the ischemic core and a marked functional recovery. The functional benefits obtained by the timely injection of the ELRs-hydrogel in a clinically relevant MI model support the potential utility of this treatment for further clinical translation.
Collapse
Affiliation(s)
- Paolo Contessotto
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Doriana Orbanić
- Group for Advanced Materials and Nanobiotechnology (BIOFORGE Lab), CIBER-BBN, University of Valladolid, Valladolid, Spain
| | - Mark Da Costa
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland.
| | - Chunsheng Jin
- Department of Medical Biochemistry, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Owens
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Sandrine Chantepie
- Laboratory Cell Growth, Tissue Repair, and Regeneration (CRRET), EA UPEC 4397/ERL CNRS 9215, University Paris Est, Créteil, France
| | - Clizia Chinello
- Clinical Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, Italy
| | - John Newell
- School of Mathematics, Statistics, and Applied Mathematics, National University of Ireland Galway, Galway, Ireland
| | - Fulvio Magni
- Clinical Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, Italy
| | - Dulce Papy-Garcia
- Laboratory Cell Growth, Tissue Repair, and Regeneration (CRRET), EA UPEC 4397/ERL CNRS 9215, University Paris Est, Créteil, France
| | - Niclas G Karlsson
- Department of Medical Biochemistry, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Michelle Kilcoyne
- Carbohydrate Signalling Group, Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Peter Dockery
- Centre for Microscopy and Imaging, Anatomy, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - José C Rodríguez-Cabello
- Group for Advanced Materials and Nanobiotechnology (BIOFORGE Lab), CIBER-BBN, University of Valladolid, Valladolid, Spain
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
15
|
Contreras O, Rossi FMV, Theret M. Origins, potency, and heterogeneity of skeletal muscle fibro-adipogenic progenitors-time for new definitions. Skelet Muscle 2021; 11:16. [PMID: 34210364 PMCID: PMC8247239 DOI: 10.1186/s13395-021-00265-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Striated muscle is a highly plastic and regenerative organ that regulates body movement, temperature, and metabolism-all the functions needed for an individual's health and well-being. The muscle connective tissue's main components are the extracellular matrix and its resident stromal cells, which continuously reshape it in embryonic development, homeostasis, and regeneration. Fibro-adipogenic progenitors are enigmatic and transformative muscle-resident interstitial cells with mesenchymal stem/stromal cell properties. They act as cellular sentinels and physiological hubs for adult muscle homeostasis and regeneration by shaping the microenvironment by secreting a complex cocktail of extracellular matrix components, diffusible cytokines, ligands, and immune-modulatory factors. Fibro-adipogenic progenitors are the lineage precursors of specialized cells, including activated fibroblasts, adipocytes, and osteogenic cells after injury. Here, we discuss current research gaps, potential druggable developments, and outstanding questions about fibro-adipogenic progenitor origins, potency, and heterogeneity. Finally, we took advantage of recent advances in single-cell technologies combined with lineage tracing to unify the diversity of stromal fibro-adipogenic progenitors. Thus, this compelling review provides new cellular and molecular insights in comprehending the origins, definitions, markers, fate, and plasticity of murine and human fibro-adipogenic progenitors in muscle development, homeostasis, regeneration, and repair.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, 2052, Australia.
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fabio M V Rossi
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
16
|
Neill T, Kapoor A, Xie C, Buraschi S, Iozzo RV. A functional outside-in signaling network of proteoglycans and matrix molecules regulating autophagy. Matrix Biol 2021; 100-101:118-149. [PMID: 33838253 PMCID: PMC8355044 DOI: 10.1016/j.matbio.2021.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Proteoglycans and selected extracellular matrix constituents are emerging as intrinsic and critical regulators of evolutionarily conversed, intracellular catabolic pathways. Often, these secreted molecules evoke sustained autophagy in a variety of cell types, tissues, and model systems. The unique properties of proteoglycans have ushered in a paradigmatic shift to broaden our understanding of matrix-mediated signaling cascades. The dynamic cellular pathway controlling autophagy is now linked to an equally dynamic and fluid signaling network embedded in a complex meshwork of matrix molecules. A rapidly emerging field of research encompasses multiple matrix-derived candidates, representing a menagerie of soluble matrix constituents including decorin, biglycan, endorepellin, endostatin, collagen VI and plasminogen kringle 5. These matrix constituents are pro-autophagic and simultaneously anti-angiogenic. In contrast, perlecan, laminin α2 chain, and lumican have anti-autophagic functions. Mechanistically, each matrix constituent linked to intracellular catabolic events engages a specific cell surface receptor that often converges on a common core of the autophagic machinery including AMPK, Peg3 and Beclin 1. We consider this matrix-evoked autophagy as non-canonical given that it occurs in an allosteric manner and is independent of nutrient availability or prevailing bioenergetics control. We propose that matrix-regulated autophagy is an important outside-in signaling mechanism for proper tissue homeostasis that could be therapeutically leveraged to combat a variety of diseases.
Collapse
Affiliation(s)
- Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Aastha Kapoor
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christopher Xie
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Simone Buraschi
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
17
|
Ausems CRM, van Engelen BGM, van Bokhoven H, Wansink DG. Systemic cell therapy for muscular dystrophies : The ultimate transplantable muscle progenitor cell and current challenges for clinical efficacy. Stem Cell Rev Rep 2021; 17:878-899. [PMID: 33349909 PMCID: PMC8166694 DOI: 10.1007/s12015-020-10100-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 01/07/2023]
Abstract
The intrinsic regenerative capacity of skeletal muscle makes it an excellent target for cell therapy. However, the potential of muscle tissue to renew is typically exhausted and insufficient in muscular dystrophies (MDs), a large group of heterogeneous genetic disorders showing progressive loss of skeletal muscle fibers. Cell therapy for MDs has to rely on suppletion with donor cells with high myogenic regenerative capacity. Here, we provide an overview on stem cell lineages employed for strategies in MDs, with a focus on adult stem cells and progenitor cells resident in skeletal muscle. In the early days, the potential of myoblasts and satellite cells was explored, but after disappointing clinical results the field moved to other muscle progenitor cells, each with its own advantages and disadvantages. Most recently, mesoangioblasts and pericytes have been pursued for muscle cell therapy, leading to a handful of preclinical studies and a clinical trial. The current status of (pre)clinical work for the most common forms of MD illustrates the existing challenges and bottlenecks. Besides the intrinsic properties of transplantable cells, we discuss issues relating to cell expansion and cell viability after transplantation, optimal dosage, and route and timing of administration. Since MDs are genetic conditions, autologous cell therapy and gene therapy will need to go hand-in-hand, bringing in additional complications. Finally, we discuss determinants for optimization of future clinical trials for muscle cell therapy. Joined research efforts bring hope that effective therapies for MDs are on the horizon to fulfil the unmet clinical need in patients.
Collapse
Affiliation(s)
- C Rosanne M Ausems
- Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
- Donders lnstitute for Brain Cognition and Behavior, Department of Neurology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
- Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Baziel G M van Engelen
- Donders lnstitute for Brain Cognition and Behavior, Department of Neurology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands
| | - Hans van Bokhoven
- Donders lnstitute for Brain Cognition and Behavior, Department of Human Genetics, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands.
| | - Derick G Wansink
- Radboud Institute for Molecular Life Sciences, Department of Cell Biology, Radboud University Medical Center, 6525, GA, Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Giacomazzi G, Giovannelli G, Rotini A, Costamagna D, Quattrocelli M, Sampaolesi M. Isolation of Mammalian Mesoangioblasts: A Subset of Pericytes with Myogenic Potential. Methods Mol Biol 2021; 2235:155-167. [PMID: 33576976 DOI: 10.1007/978-1-0716-1056-5_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mesoangioblasts (MABs) are vessel-associated stem cells that express pericyte markers and are originally isolated from the embryonic dorsal aorta. From postnatal small vessels of skeletal muscle and heart, it is possible to isolate cells with similar characteristics to embryonic MABs. Adult MABs have the capacity to self-renew and to differentiate into cell types of mesodermal lineages upon proper culture conditions. To date, the origin of MABs and the relationship with other muscle stem cells are still debated. Recently, in a phase I-II clinical trial, intra-arterial HLA-matched MABs were proved to be relatively safe. Novel information on MAB pure populations is desirable, and implementation of their therapeutic potential is mandatory to approach efficacy in MAB-based treatments. This chapter provides an overview of the current techniques for isolation and characterization of rodent, canine, human, and equine adult MABs.
Collapse
Affiliation(s)
- Giorgia Giacomazzi
- Translational Cardiomyology Laboratory, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Gaia Giovannelli
- Translational Cardiomyology Laboratory, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Neuroscience Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Alessio Rotini
- Translational Cardiomyology Laboratory, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Neuroscience Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Domiziana Costamagna
- Translational Cardiomyology Laboratory, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Mattia Quattrocelli
- Translational Cardiomyology Laboratory, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
19
|
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
|
20
|
Wang C, Xu X, Chen J, Kang Y, Guo J, Duscher D, Yang X, Guo G, Ren S, Xiong H, Yuan M, Jiang T, Machens HG, Chen Z, Chen Y. The Construction and Analysis of lncRNA-miRNA-mRNA Competing Endogenous RNA Network of Schwann Cells in Diabetic Peripheral Neuropathy. Front Bioeng Biotechnol 2020; 8:490. [PMID: 32523943 PMCID: PMC7261901 DOI: 10.3389/fbioe.2020.00490] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background Diabetes mellitus is a worldwide disease with high incidence. Diabetic peripheral neuropathy (DPN) is one of the most common but often ignored complications of diabetes mellitus that cause numbness and pain, even paralysis. Recent studies demonstrate that Schwann cells (SCs) in the peripheral nervous system play an essential role in the pathogenesis of DPN. Furthermore, various transcriptome analyses constructed by RNA-seq or microarray have provided a comprehensive understanding of molecular mechanisms and regulatory interaction networks involved in many diseases. However, the detailed mechanisms and competing endogenous RNA (ceRNA) network of SCs in DPN remain largely unknown. Methods Whole-transcriptome sequencing technology was applied to systematically analyze the differentially expressed mRNAs, lncRNAs and miRNAs in SCs from DPN rats and control rats. Gene ontology (GO) and KEGG pathway enrichment analyses were used to investigate the potential functions of the differentially expressed genes. Following this, lncRNA-mRNA co-expression network and ceRNA regulatory network were constructed by bioinformatics analysis methods. Results The results showed that 2925 mRNAs, 164 lncRNAs and 49 miRNAs were significantly differently expressed in SCs from DPN rats compared with control rats. 13 mRNAs, 7 lncRNAs and 7 miRNAs were validated by qRT-PCR and consistent with the RNA-seq data. Functional and pathway analyses revealed that many enriched biological processes of GO terms and pathways were highly correlated with the function of SCs and the pathogenesis of DPN. Furthermore, a global lncRNA–miRNA–mRNA ceRNA regulatory network in DPN model was constructed and miR-212-5p and the significantly correlated lncRNAs with high degree were identified as key mediators in the pathophysiological processes of SCs in DPN. These RNAs would contribute to the diagnosis and treatment of DPN. Conclusion Our study has shown that differentially expressed RNAs have complex interactions among them. They also play critical roles in regulating functions of SCs involved in the pathogenesis of DPN. The novel competitive endogenous RNA network provides new insight for exploring the underlying molecular mechanism of DPN and further investigation may have clinical application value.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahe Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dominik Duscher
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich, Germany
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guojun Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sen Ren
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hewei Xiong
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Yuan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich, Germany
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhua Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Teng K, Luan X, An Q, Zhao Y, Hu X, Zhang S, Zhuang J, Li X, Lu L, Zhang Y. Orthogonally Regulated Mechanical Strength and Molecular Delivery Capabilities Achieved in a Double Network Hydrogel Matrix. ChemistrySelect 2020. [DOI: 10.1002/slct.202000620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Kaixuan Teng
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Xinglong Luan
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Qi An
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Yantao Zhao
- Beijing Engineering Research Center of Orthopaedic Implants Fourth Medical Center of CPLA General Hospital Beijing 100048 China
| | - Xiantong Hu
- Beijing Engineering Research Center of Orthopaedic Implants Fourth Medical Center of CPLA General Hospital Beijing 100048 China
| | - Shuting Zhang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Jialin Zhuang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Xiaobo Li
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Limei Lu
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| | - Yihe Zhang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes National Laboratory of Mineral Materials School of Materials Sciences and Technology China University of Geosciences Beijing 100083 China
| |
Collapse
|
22
|
Zeb2 Regulates Myogenic Differentiation in Pluripotent Stem Cells. Int J Mol Sci 2020; 21:ijms21072525. [PMID: 32260521 PMCID: PMC7177401 DOI: 10.3390/ijms21072525] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle differentiation is triggered by a unique family of myogenic basic helix-loop-helix transcription factors, including MyoD, MRF-4, Myf-5, and Myogenin. These transcription factors bind promoters and distant regulatory regions, including E-box elements, of genes whose expression is restricted to muscle cells. Other E-box binding zinc finger proteins target the same DNA response elements, however, their function in muscle development and regeneration is still unknown. Here, we show that the transcription factor zinc finger E-box-binding homeobox 2 (Zeb2, Sip-1, Zfhx1b) is present in skeletal muscle tissues. We investigate the role of Zeb2 in skeletal muscle differentiation using genetic tools and transgenic mouse embryonic stem cells, together with single-cell RNA-sequencing and in vivo muscle engraftment capability. We show that Zeb2 over-expression has a positive impact on skeletal muscle differentiation in pluripotent stem cells and adult myogenic progenitors. We therefore propose that Zeb2 is a novel myogenic regulator and a possible target for improving skeletal muscle regeneration. The non-neural roles of Zeb2 are poorly understood.
Collapse
|
23
|
Mavoungou LO, Neuenschwander S, Pham U, Iyer PS, Mermod N. Characterization of mesoangioblast cell fate and improved promyogenic potential of a satellite cell-like subpopulation upon transplantation in dystrophic murine muscles. Stem Cell Res 2019; 41:101619. [PMID: 31683098 DOI: 10.1016/j.scr.2019.101619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/20/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease caused by the lack of dystrophin in muscle fibers that is currently without curative treatment. Mesoangioblasts (MABs) are multipotent progenitor cells that can differentiate to a myogenic lineage and that can be used to express Dystrophin upon transplantation into muscles, in autologous gene therapy approaches. However, their fate in the muscle environment remains poorly characterized. Here, we investigated the differentiation fate of MABs following their transplantation in DMD murine muscles using a mass cytometry strategy. This allowed the identification and isolation of a fraction of MAB-derived cells presenting common properties with satellite muscle stem cells. This analysis also indicated that most cells did not undergo a myogenic differentiation path once in the muscle environment, limiting their capacity to restore dystrophin expression in transplanted muscles. We therefore assessed whether MAB treatment with cytokines and growth factors prior to engraftment may improve their myogenic fate. We identified a combination of such signals that ameliorates MABs capacity to undergo myogenic differentiation in vivo and to restore dystrophin expression upon engraftment in myopathic murine muscles.
Collapse
Affiliation(s)
- Lionel O Mavoungou
- Institute of Biotechnology and Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | | | - Uyen Pham
- Grand Valley State University, MI, USA
| | - Pavithra S Iyer
- Institute of Biotechnology and Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland; Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zürich, Switzerland
| | - Nicolas Mermod
- Institute of Biotechnology and Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
24
|
Huang M, Chen Y, Han D, Lei Z, Chu X. Role of the zinc finger and SCAN domain-containing transcription factors in cancer. Am J Cancer Res 2019; 9:816-836. [PMID: 31218096 PMCID: PMC6556609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 04/04/2019] [Indexed: 06/09/2023] Open
Abstract
Transcription factors are key determinants of gene expression that recognize and bind to short DNA sequence motifs, thereby regulating many biological processes including differentiation, development, and metabolism. Transcription factors are increasingly recognized for their roles in cancer progression. Here, we describe a subfamily of zinc finger transcription factors named zinc finger and SCAN domain containing (ZSCAN) transcription factors. In this review, we summarize the identified members of the ZSCAN family of transcription factors and their roles in cancer progression. Due to the complex regulation mechanisms, ZSCAN transcription factors may show promotive or prohibitive efforts in angiogenesis, cell apoptosis, cell differentiation, cell migration and invasion, cell proliferation, stem cell properties, and chemotherapy sensitivity. The upstream regulation mechanisms of their varied expression levels may include gene mutation, DNA methylation, alternative splicing, and miRNA regulation. What's more, to clarify their diverse functions, we summarize the modulation mechanisms of their activity in downstream genes transcription, including protein-protein interactions mediated by their SCAN box, recruitment of co-regulating molecules and post-translational modifications. A better understanding of the widespread regulatory mode of these transcription factors will provide further insight into the mechanism of transcriptional regulation and suggest novel therapeutic strategies against tumor progression.
Collapse
Affiliation(s)
- Mengxi Huang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu Province, People’s Republic of China
| | - Yanyan Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu Province, People’s Republic of China
| | - Dong Han
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, People’s Republic of China
| | - Zengjie Lei
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu Province, People’s Republic of China
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, People’s Republic of China
| | - Xiaoyuan Chu
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing UniversityNanjing, Jiangsu Province, People’s Republic of China
- Department of Medical Oncology, Jinling Hospital, Nanjing Clinical School of Southern Medical UniversityNanjing, Jiangsu Province, People’s Republic of China
| |
Collapse
|
25
|
Chiappalupi S, Salvadori L, Luca G, Riuzzi F, Calafiore R, Donato R, Sorci G. Do porcine Sertoli cells represent an opportunity for Duchenne muscular dystrophy? Cell Prolif 2019; 52:e12599. [PMID: 30912260 PMCID: PMC6536415 DOI: 10.1111/cpr.12599] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/24/2019] [Accepted: 02/09/2019] [Indexed: 12/13/2022] Open
Abstract
Sertoli cells (SeC) are responsible for the immunoprivileged status of the testis thanks to which allogeneic or xenogeneic engraftments can survive without pharmacological immune suppression if co‐injected with SeC. This peculiar ability of SeC is dependent on secretion of a plethora of factors including maturation factors, hormones, growth factors, cytokines and immunomodulatory factors. The anti‐inflammatory and trophic properties of SeC have been largely exploited in several experimental models of diseases, diabetes being the most studied. Duchenne muscular dystrophy (DMD) is a lethal X‐linked recessive pathology in which lack of functional dystrophin leads to progressive muscle degeneration culminating in loss of locomotion and premature death. Despite a huge effort to find a cure, DMD patients are currently treated with anti‐inflammatory steroids. Recently, encapsulated porcine SeC (MC‐SeC) have been injected ip in the absence of immunosuppression in an animal model of DMD resulting in reduction of muscle inflammation and amelioration of muscle morphology and functionality, thus opening an additional avenue in the treatment of DMD. The novel protocol is endowed with the advantage of being potentially applicable to all the cohort of DMD patients regardless of the mutation. This mini‐review addresses several issues linked to the possible use of MC‐SeC injected ip in dystrophic people.
Collapse
Affiliation(s)
- Sara Chiappalupi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy
| | - Laura Salvadori
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Francesca Riuzzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy
| | | | - Rosario Donato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, Italy
| |
Collapse
|
26
|
Teng S, Huang P. The effect of type 2 diabetes mellitus and obesity on muscle progenitor cell function. Stem Cell Res Ther 2019; 10:103. [PMID: 30898146 PMCID: PMC6427880 DOI: 10.1186/s13287-019-1186-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In addition to its primary function to provide movement and maintain posture, the skeletal muscle plays important roles in energy and glucose metabolism. In healthy humans, skeletal muscle is the major site for postprandial glucose uptake and impairment of this process contributes to the pathogenesis of type 2 diabetes mellitus (T2DM). A key component to the maintenance of skeletal muscle integrity and plasticity is the presence of muscle progenitor cells, including satellite cells, fibroadipogenic progenitors, and some interstitial progenitor cells associated with vessels (myo-endothelial cells, pericytes, and mesoangioblasts). In this review, we aim to discuss the emerging concepts related to these progenitor cells, focusing on the identification and characterization of distinct progenitor cell populations, and the impact of obesity and T2DM on these cells. The recent advances in stem cell therapies by targeting diabetic and obese muscle are also discussed.
Collapse
Affiliation(s)
- Shuzhi Teng
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, 126 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China.
| | - Ping Huang
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, 126 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China.
| |
Collapse
|
27
|
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
|
28
|
Abstract
Skeletal muscle regeneration is a highly orchestrated process and involves the activation of many cellular and molecular pathways. Although satellite cells (SCs) are the major cell type responsible for muscle regeneration, pericytes show remarkable myogenic potential and various advantages as cell therapy in muscular disorders. This chapter first introduces the structure, marker expression, origin, and category of pericytes. Next, we discuss their functions in muscular dystrophy and/or muscle injuries, focusing on their myogenic, adipogenic, fibrogenic, chondrogenic, and osteogenic activities. Understanding this knowledge will promote the development of innovative cell therapies for muscle disorders, including muscular dystrophy.
Collapse
|
29
|
Evano B, Tajbakhsh S. Skeletal muscle stem cells in comfort and stress. NPJ Regen Med 2018; 3:24. [PMID: 30588332 PMCID: PMC6303387 DOI: 10.1038/s41536-018-0062-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
Investigations on developmental and regenerative myogenesis have led to major advances in decrypting stem cell properties and potential, as well as their interactions within the evolving niche. As a consequence, regenerative myogenesis has provided a forum to investigate intrinsic regulators of stem cell properties as well as extrinsic factors, including stromal cells, during normal growth and following injury and disease. Here we review some of the latest advances in the field that have exposed fundamental processes including regulation of stress following trauma and ageing, senescence, DNA damage control and modes of symmetric and asymmetric cell divisions. Recent studies have begun to explore the nature of the niche that is distinct in different muscle groups, and that is altered from prenatal to postnatal stages, and during ageing. We also discuss heterogeneities among muscle stem cells and how distinct properties within the quiescent and proliferating cell states might impact on homoeostasis and regeneration. Interestingly, cellular quiescence, which was thought to be a passive cell state, is regulated by multiple mechanisms, many of which are deregulated in various contexts including ageing. These and other factors including metabolic activity and genetic background can impact on the efficiency of muscle regeneration.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 75015 Paris, France
- CNRS UMR 3738, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
30
|
Correra RM, Ollitrault D, Valente M, Mazzola A, Adalsteinsson BT, Ferguson-Smith AC, Marazzi G, Sassoon DA. The imprinted gene Pw1/Peg3 regulates skeletal muscle growth, satellite cell metabolic state, and self-renewal. Sci Rep 2018; 8:14649. [PMID: 30279563 PMCID: PMC6168517 DOI: 10.1038/s41598-018-32941-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/18/2018] [Indexed: 12/16/2022] Open
Abstract
Pw1/Peg3 is an imprinted gene expressed from the paternally inherited allele. Several imprinted genes, including Pw1/Peg3, have been shown to regulate overall body size and play a role in adult stem cells. Pw1/Peg3 is expressed in muscle stem cells (satellite cells) as well as a progenitor subset of muscle interstitial cells (PICs) in adult skeletal muscle. We therefore examined the impact of loss-of-function of Pw1/Peg3 during skeletal muscle growth and in muscle stem cell behavior. We found that constitutive loss of Pw1/Peg3 function leads to a reduced muscle mass and myofiber number. In newborn mice, the reduction in fiber number is increased in homozygous mutants as compared to the deletion of only the paternal Pw1/Peg3 allele, indicating that the maternal allele is developmentally functional. Constitutive and a satellite cell-specific deletion of Pw1/Peg3, revealed impaired muscle regeneration and a reduced capacity of satellite cells for self-renewal. RNA sequencing analyses revealed a deregulation of genes that control mitochondrial function. Consistent with these observations, Pw1/Peg3 mutant satellite cells displayed increased mitochondrial activity coupled with accelerated proliferation and differentiation. Our data show that Pw1/Peg3 regulates muscle fiber number determination during fetal development in a gene-dosage manner and regulates satellite cell metabolism in the adult.
Collapse
Affiliation(s)
- Rosa Maria Correra
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
| | - David Ollitrault
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Mariana Valente
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Alessia Mazzola
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
| | - Bjorn T Adalsteinsson
- Department of Physiology Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, United Kingdom
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - Giovanna Marazzi
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France.
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France.
| | - David A Sassoon
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France.
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France.
| |
Collapse
|
31
|
Slukvin II, Kumar A. The mesenchymoangioblast, mesodermal precursor for mesenchymal and endothelial cells. Cell Mol Life Sci 2018; 75:3507-3520. [PMID: 29992471 PMCID: PMC6328351 DOI: 10.1007/s00018-018-2871-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/29/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Mesenchymoangioblast (MB) is the earliest precursor for endothelial and mesenchymal cells originating from APLNR+PDGFRα+KDR+ mesoderm in human pluripotent stem cell cultures. MBs are identified based on their capacity to form FGF2-dependent compact spheroid colonies in a serum-free semisolid medium. MBs colonies are composed of PDGFRβ+CD271+EMCN+DLK1+CD73- primitive mesenchymal cells which are generated through endothelial/angioblastic intermediates (cores) formed during first 3-4 days of clonogenic cultures. MB-derived primitive mesenchymal cells have potential to differentiate into mesenchymal stromal/stem cells (MSCs), pericytes, and smooth muscle cells. In this review, we summarize the specification and developmental potential of MBs, emphasize features that distinguish MBs from other mesenchymal progenitors described in the literature and discuss the value of these findings for identifying molecular pathways leading to MSC and vasculogenic cell specification, and developing cellular therapies using MB-derived progeny.
Collapse
Affiliation(s)
- Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin, 1220 Capitol Ct., Madison, WI, 53715, USA.
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53707, USA.
- Department of Pathology and Laboratory Medicine, University of Wisconsin, 1685 Highland Ave, Madison, WI, 53705, USA.
| | - Akhilesh Kumar
- Wisconsin National Primate Research Center, University of Wisconsin, 1220 Capitol Ct., Madison, WI, 53715, USA
| |
Collapse
|
32
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [PMID: 30204432 DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
33
|
André LM, Ausems CRM, Wansink DG, Wieringa B. Abnormalities in Skeletal Muscle Myogenesis, Growth, and Regeneration in Myotonic Dystrophy. Front Neurol 2018; 9:368. [PMID: 29892259 PMCID: PMC5985300 DOI: 10.3389/fneur.2018.00368] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/07/2018] [Indexed: 12/16/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) and 2 (DM2) are autosomal dominant degenerative neuromuscular disorders characterized by progressive skeletal muscle weakness, atrophy, and myotonia with progeroid features. Although both DM1 and DM2 are characterized by skeletal muscle dysfunction and also share other clinical features, the diseases differ in the muscle groups that are affected. In DM1, distal muscles are mainly affected, whereas in DM2 problems are mostly found in proximal muscles. In addition, manifestation in DM1 is generally more severe, with possible congenital or childhood-onset of disease and prominent CNS involvement. DM1 and DM2 are caused by expansion of (CTG•CAG)n and (CCTG•CAGG)n repeats in the 3' non-coding region of DMPK and in intron 1 of CNBP, respectively, and in overlapping antisense genes. This critical review will focus on the pleiotropic problems that occur during development, growth, regeneration, and aging of skeletal muscle in patients who inherited these expansions. The current best-accepted idea is that most muscle symptoms can be explained by pathomechanistic effects of repeat expansion on RNA-mediated pathways. However, aberrations in DNA replication and transcription of the DM loci or in protein translation and proteome homeostasis could also affect the control of proliferation and differentiation of muscle progenitor cells or the maintenance and physiological integrity of muscle fibers during a patient's lifetime. Here, we will discuss these molecular and cellular processes and summarize current knowledge about the role of embryonic and adult muscle-resident stem cells in growth, homeostasis, regeneration, and premature aging of healthy and diseased muscle tissue. Of particular interest is that also progenitor cells from extramuscular sources, such as pericytes and mesoangioblasts, can participate in myogenic differentiation. We will examine the potential of all these types of cells in the application of regenerative medicine for muscular dystrophies and evaluate new possibilities for their use in future therapy of DM.
Collapse
Affiliation(s)
- Laurène M André
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - C Rosanne M Ausems
- Department of Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bé Wieringa
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
34
|
Iyer PS, Mavoungou LO, Ronzoni F, Zemla J, Schmid-Siegert E, Antonini S, Neff LA, Dorchies OM, Jaconi M, Lekka M, Messina G, Mermod N. Autologous Cell Therapy Approach for Duchenne Muscular Dystrophy using PiggyBac Transposons and Mesoangioblasts. Mol Ther 2018; 26:1093-1108. [PMID: 29503200 PMCID: PMC6079556 DOI: 10.1016/j.ymthe.2018.01.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 01/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease currently without cure. We investigated the use of the PiggyBac transposon for full-length dystrophin expression in murine mesoangioblast (MABs) progenitor cells. DMD murine MABs were transfected with transposable expression vectors for full-length dystrophin and transplanted intramuscularly or intra-arterially into mdx/SCID mice. Intra-arterial delivery indicated that the MABs could migrate to regenerating muscles to mediate dystrophin expression. Intramuscular transplantation yielded dystrophin expression in 11%-44% of myofibers in murine muscles, which remained stable for the assessed period of 5 months. The satellite cells isolated from transplanted muscles comprised a fraction of MAB-derived cells, indicating that the transfected MABs may colonize the satellite stem cell niche. Transposon integration site mapping by whole-genome sequencing indicated that 70% of the integrations were intergenic, while none was observed in an exon. Muscle resistance assessment by atomic force microscopy indicated that 80% of fibers showed elasticity properties restored to those of wild-type muscles. As measured in vivo, transplanted muscles became more resistant to fatigue. This study thus provides a proof-of-principle that PiggyBac transposon vectors may mediate full-length dystrophin expression as well as functional amelioration of the dystrophic muscles within a potential autologous cell-based therapeutic approach of DMD.
Collapse
Affiliation(s)
- Pavithra S Iyer
- Institute of Biotechnology, University of Lausanne, Lausanne, Switzerland
| | - Lionel O Mavoungou
- Institute of Biotechnology, University of Lausanne, Lausanne, Switzerland
| | - Flavio Ronzoni
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Joanna Zemla
- Institute of Nuclear Physics, Polish Academy of Sciences, 31342 Krakow, Poland
| | | | | | - Laurence A Neff
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Olivier M Dorchies
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, 1211 Geneva, Switzerland
| | - Marisa Jaconi
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Malgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, 31342 Krakow, Poland
| | | | - Nicolas Mermod
- Institute of Biotechnology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
35
|
Čamernik K, Barlič A, Drobnič M, Marc J, Jeras M, Zupan J. Mesenchymal Stem Cells in the Musculoskeletal System: From Animal Models to Human Tissue Regeneration? Stem Cell Rev Rep 2018; 14:346-369. [DOI: 10.1007/s12015-018-9800-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Shafiee A, Patel J, Hutmacher DW, Fisk NM, Khosrotehrani K. Meso-Endothelial Bipotent Progenitors from Human Placenta Display Distinct Molecular and Cellular Identity. Stem Cell Reports 2018; 10:890-904. [PMID: 29478891 PMCID: PMC5918195 DOI: 10.1016/j.stemcr.2018.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
The existence of bipotential precursors for both mesenchymal and endothelial stem/progenitor cells in human postnatal life is debated. Here, we hypothesized that such progenitors are present within the human term placenta. From a heterogeneous placental single-cell suspension, a directly flow-sorted CD45-CD34+CD144+CD31Lo population uniquely differentiated into both endothelial and mesenchymal colonies in limiting dilution culture assays. Of interest, these bipotent cells were in vessel walls but not in contact with the circulation. RNA sequencing and functional analysis demonstrated that Notch signaling was a key driver for endothelial and bipotential progenitor function. In contrast, the formation of mesenchymal cells from the bipotential population was not affected by TGFβ receptor inhibition, a classical pathway for endothelial-mesenchymal transition. This study reveals a bipotent progenitor phenotype in the human placenta at the cellular and molecular levels, giving rise to endothelial and mesenchymal cells ex vivo.
Collapse
Affiliation(s)
- Abbas Shafiee
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia; UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jatin Patel
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicholas M Fisk
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; Faculty of Medicine, UNSW, Sydney, NSW, Australia; Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, QLD, Australia
| | - Kiarash Khosrotehrani
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
37
|
Benedetti S, Uno N, Hoshiya H, Ragazzi M, Ferrari G, Kazuki Y, Moyle LA, Tonlorenzi R, Lombardo A, Chaouch S, Mouly V, Moore M, Popplewell L, Kazuki K, Katoh M, Naldini L, Dickson G, Messina G, Oshimura M, Cossu G, Tedesco FS. Reversible immortalisation enables genetic correction of human muscle progenitors and engineering of next-generation human artificial chromosomes for Duchenne muscular dystrophy. EMBO Mol Med 2018; 10:254-275. [PMID: 29242210 PMCID: PMC5801502 DOI: 10.15252/emmm.201607284] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 11/07/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022] Open
Abstract
Transferring large or multiple genes into primary human stem/progenitor cells is challenged by restrictions in vector capacity, and this hurdle limits the success of gene therapy. A paradigm is Duchenne muscular dystrophy (DMD), an incurable disorder caused by mutations in the largest human gene: dystrophin. The combination of large-capacity vectors, such as human artificial chromosomes (HACs), with stem/progenitor cells may overcome this limitation. We previously reported amelioration of the dystrophic phenotype in mice transplanted with murine muscle progenitors containing a HAC with the entire dystrophin locus (DYS-HAC). However, translation of this strategy to human muscle progenitors requires extension of their proliferative potential to withstand clonal cell expansion after HAC transfer. Here, we show that reversible cell immortalisation mediated by lentivirally delivered excisable hTERT and Bmi1 transgenes extended cell proliferation, enabling transfer of a novel DYS-HAC into DMD satellite cell-derived myoblasts and perivascular cell-derived mesoangioblasts. Genetically corrected cells maintained a stable karyotype, did not undergo tumorigenic transformation and retained their migration ability. Cells remained myogenic in vitro (spontaneously or upon MyoD induction) and engrafted murine skeletal muscle upon transplantation. Finally, we combined the aforementioned functions into a next-generation HAC capable of delivering reversible immortalisation, complete genetic correction, additional dystrophin expression, inducible differentiation and controllable cell death. This work establishes a novel platform for complex gene transfer into clinically relevant human muscle progenitors for DMD gene therapy.
Collapse
Affiliation(s)
- Sara Benedetti
- Department of Cell and Developmental Biology, University College London, London, UK
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Narumi Uno
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Tottori University, Yonago, Tottori, Japan
- Chromosome Engineering Research Center (CERC), Tottori University, Yonago, Tottori, Japan
| | - Hidetoshi Hoshiya
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Giulia Ferrari
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Yasuhiro Kazuki
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Tottori University, Yonago, Tottori, Japan
- Chromosome Engineering Research Center (CERC), Tottori University, Yonago, Tottori, Japan
| | - Louise Anne Moyle
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Rossana Tonlorenzi
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Lombardo
- San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute and Vita Salute San Raffaele University, Milan, Italy
| | - Soraya Chaouch
- AIM/AFM Center for Research in Myology, Sorbonne Universités, UPMC Univ. Paris 06, INSERM UMRS974, CNRS FRE3617, Paris, France
| | - Vincent Mouly
- AIM/AFM Center for Research in Myology, Sorbonne Universités, UPMC Univ. Paris 06, INSERM UMRS974, CNRS FRE3617, Paris, France
| | - Marc Moore
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey, UK
| | - Linda Popplewell
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey, UK
| | - Kanako Kazuki
- Chromosome Engineering Research Center (CERC), Tottori University, Yonago, Tottori, Japan
| | - Motonobu Katoh
- Chromosome Engineering Research Center (CERC), Tottori University, Yonago, Tottori, Japan
| | - Luigi Naldini
- Department of Biosciences, University of Milan, Milan, Italy
| | - George Dickson
- School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey, UK
| | | | - Mitsuo Oshimura
- Chromosome Engineering Research Center (CERC), Tottori University, Yonago, Tottori, Japan
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | | |
Collapse
|
38
|
Fischer T, Wilharm N, Hayn A, Mierke CT. Matrix and cellular mechanical properties are the driving factors for facilitating human cancer cell motility into 3D engineered matrices. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa8bbb] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
39
|
Yin YJ, Huang YP, Lu C, Sun XP, Niu FN, Jin R, Zhou GP. [A retrospective analysis of 6 children with Duchenne muscular dystrophy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:405-409. [PMID: 28407826 PMCID: PMC7389655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/28/2016] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To analyze the clinical features of 6 children with Duchenne muscular dystrophy (DMD) and review related literature, and to provide a basis for early diagnosis and effective treatment of this disease. METHODS A retrospective analysis was performed on the clinical data of 6 children with DMD who were admitted to the First Affiliated Hospital of Nanjing Medical University from January 2010 to October 2015. RESULTS All the 6 cases were boys without a family history of DMD, and the age of diagnosis of DMD was 1.2-11.5 years. All patients had insidious onset and increases in alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, α-hydroxybutyrate dehydrogenase, creatine kinase (CK), and creatine kinase-MB, particularly CK, which was 3.3-107.2 times the normal level. Their gene detection results all showed DMD gene mutation. The gene detection results of two children's mothers showed that they carried the same mutant gene. The muscle biopsy in one case showed that the pathological changes confirmed the diagnosis of DMD. The level of CK in one case declined by 77.0% 5 days after umbilical cord blood mesenchymal stem cell transplantation. CONCLUSIONS For boys with abnormal serum enzyme levels and motor function, DMD should be highly suspected. It should be confirmed by CK and DMD gene detection as soon as possible. And the progression of the disease could be delayed by early intervention for protecting the remaining normal muscle fibers.
Collapse
Affiliation(s)
- Yu-Jie Yin
- Department of Pediatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | | | | | | | | | | | | |
Collapse
|
40
|
Yin YJ, Huang YP, Lu C, Sun XP, Niu FN, Jin R, Zhou GP. [A retrospective analysis of 6 children with Duchenne muscular dystrophy]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:405-409. [PMID: 28407826 PMCID: PMC7389655 DOI: 10.7499/j.issn.1008-8830.2017.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/28/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To analyze the clinical features of 6 children with Duchenne muscular dystrophy (DMD) and review related literature, and to provide a basis for early diagnosis and effective treatment of this disease. METHODS A retrospective analysis was performed on the clinical data of 6 children with DMD who were admitted to the First Affiliated Hospital of Nanjing Medical University from January 2010 to October 2015. RESULTS All the 6 cases were boys without a family history of DMD, and the age of diagnosis of DMD was 1.2-11.5 years. All patients had insidious onset and increases in alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, α-hydroxybutyrate dehydrogenase, creatine kinase (CK), and creatine kinase-MB, particularly CK, which was 3.3-107.2 times the normal level. Their gene detection results all showed DMD gene mutation. The gene detection results of two children's mothers showed that they carried the same mutant gene. The muscle biopsy in one case showed that the pathological changes confirmed the diagnosis of DMD. The level of CK in one case declined by 77.0% 5 days after umbilical cord blood mesenchymal stem cell transplantation. CONCLUSIONS For boys with abnormal serum enzyme levels and motor function, DMD should be highly suspected. It should be confirmed by CK and DMD gene detection as soon as possible. And the progression of the disease could be delayed by early intervention for protecting the remaining normal muscle fibers.
Collapse
Affiliation(s)
- Yu-Jie Yin
- Department of Pediatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | | | | | | | | | | | | |
Collapse
|
41
|
Malinverno M, Corada M, Ferrarini L, Formicola L, Marazzi G, Sassoon D, Dejana E. Peg3/PW1 Is a Marker of a Subset of Vessel Associated Endothelial Progenitors. Stem Cells 2017; 35:1328-1340. [PMID: 28090691 DOI: 10.1002/stem.2566] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/30/2016] [Accepted: 12/12/2016] [Indexed: 12/16/2022]
Abstract
Vascular associated endothelial cell (ECs) progenitors are still poorly studied and their role in the newly forming vasculature at embryonic or postnatal stage remains elusive. In the present work, we first defined a set of genes highly expressed during embryo development and strongly downregulated in the adult mouse. In this group, we then concentrated on the progenitor cell marker Peg3/PW1. By in vivo staining of the vasculature we found that only a subset of cells coexpressed endothelial markers and PW1. These cells were quite abundant in the embryo vasculature but declined in number at postnatal and adult stages. Using a reporter mouse for PW1 expression, we have been able to isolate PW1-positive (PW1posECs) and negative endothelial cells (PW1negECs). PW1-positive cells were highly proliferative in comparison to PW1negECs and were able to form colonies when seeded at clonal dilution. Furthermore, by RNAseq analysis, PW1posECs expressed endothelial cell markers together with mesenchymal and stem cell markers. When challenged by endothelial growth factors in vitro, PW1posECs were able to proliferate more than PW1negECs and to efficiently form new vessels in vivo. Taken together these data identify a subset of vessel associated endothelial cells with characteristics of progenitor cells. Considering their high proliferative potential these cells may be of particular importance to design therapies to improve the perfusion of ischemic tissues or to promote vascular repair. Stem Cells 2017;35:1328-1340.
Collapse
Affiliation(s)
- Matteo Malinverno
- FIRC Institute of Molecular Oncology (IFOM) Fondazione, Milan, Italy
| | - Monica Corada
- FIRC Institute of Molecular Oncology (IFOM) Fondazione, Milan, Italy
| | - Luca Ferrarini
- FIRC Institute of Molecular Oncology (IFOM) Fondazione, Milan, Italy
| | - Luigi Formicola
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN) UMRS 1166 Université de Pierre et Marie Curie-Sorbonne Universités and INSERM, Paris, France
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN) UMRS 1166 Université de Pierre et Marie Curie-Sorbonne Universités and INSERM, Paris, France
| | - David Sassoon
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN) UMRS 1166 Université de Pierre et Marie Curie-Sorbonne Universités and INSERM, Paris, France
| | - Elisabetta Dejana
- FIRC Institute of Molecular Oncology (IFOM) Fondazione, Milan, Italy.,Department of Biosciences, School of Sciences and Department of Oncology, School of Medicine, Milan University, Milan, Italy.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
42
|
Tonlorenzi R, Rossi G, Messina G. Isolation and Characterization of Vessel-Associated Stem/Progenitor Cells from Skeletal Muscle. Methods Mol Biol 2017; 1556:149-177. [PMID: 28247349 DOI: 10.1007/978-1-4939-6771-1_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
More than 10 years ago, we isolated from mouse embryonic dorsal aorta a population of vessel-associated stem/progenitor cells, originally named mesoangioblasts (MABs ) , capable to differentiate in all mesodermal-derived tissues, including skeletal muscle. Similar though not identical cells have been later isolated and characterized from small vessels of adult mouse and human skeletal muscles. When delivered through the arterial circulation, MABs cross the blood vessel wall and participate in skeletal muscle regeneration , leading to an amelioration of muscular dystrophies in different preclinical animal models. As such, human MABs have been used under clinical-grade conditions for a Phase I/II clinical trial for Duchenne muscular dystrophy , just concluded. Although some pericyte markers can be used to identify mouse and human MABs , no single unequivocal marker can be used to isolate MABs . As a result, MABs are mainly defined by their isolation method and functional properties. This chapter provides detailed methods for isolation, culture, and characterization of MABs in light of the recent identification of a new marker, PW1 /Peg3, to screen and identify competent MABs before their use in cell therapy.
Collapse
Affiliation(s)
- Rossana Tonlorenzi
- INSPE (Institute of Experimental Neurology) San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milano, Italy
| | - Giuliana Rossi
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
43
|
Tedesco FS, Moyle LA, Perdiguero E. Muscle Interstitial Cells: A Brief Field Guide to Non-satellite Cell Populations in Skeletal Muscle. Methods Mol Biol 2017; 1556:129-147. [PMID: 28247348 DOI: 10.1007/978-1-4939-6771-1_7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Skeletal muscle regeneration is mainly enabled by a population of adult stem cells known as satellite cells. Satellite cells have been shown to be indispensable for adult skeletal muscle repair and regeneration. In the last two decades, other stem/progenitor cell populations resident in the skeletal muscle interstitium have been identified as "collaborators" of satellite cells during regeneration. They also appear to have a key role in replacing skeletal muscle with adipose, fibrous, or bone tissue in pathological conditions. Here, we review the role and known functions of these different interstitial skeletal muscle cell types and discuss their role in skeletal muscle tissue homeostasis, regeneration, and disease, including their therapeutic potential for cell transplantation protocols.
Collapse
Affiliation(s)
- Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6DE, London, UK.
| | - Louise A Moyle
- Department of Cell and Developmental Biology, University College London, Gower Street, WC1E 6DE, London, UK
| | - Eusebio Perdiguero
- Cell Biology Group, Department of Experimental and Health Sciences (DCEXS), Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), 08003, Barcelona, Spain.
| |
Collapse
|
44
|
Besson V, Kyryachenko S, Janich P, Benitah SA, Marazzi G, Sassoon D. Expression Analysis of the Stem Cell Marker Pw1/Peg3 Reveals a CD34 Negative Progenitor Population in the Hair Follicle. Stem Cells 2016; 35:1015-1027. [PMID: 27862634 DOI: 10.1002/stem.2540] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/10/2016] [Accepted: 10/16/2016] [Indexed: 12/30/2022]
Abstract
Pw1/Peg3 is a parentally imprinted gene expressed in adult stem cells in every tissue thus far examined including the stem cells of the hair follicle. Using a Pw1/Peg3 reporter mouse, we carried out a detailed dissection of the stem cells in the bulge, which is a major stem cell compartment of the hair follicle in mammalian skin. We observed that PW1/Peg3 expression initiates upon placode formation during fetal development, coincident with the establishment of the bulge stem cells. In the adult, we observed that PW1/Peg3 expression is found in both CD34+ and CD34- populations of bulge stem cells. We demonstrate that both populations can give rise to new hair follicles, reconstitute their niche, and self-renew. These results demonstrate that PW1/Peg3 is a reliable marker of the full population of follicle stem cells and reveal a novel CD34- bulge stem-cell population. Stem Cells 2017;35:1015-1027.
Collapse
Affiliation(s)
- Vanessa Besson
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Sergiy Kyryachenko
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Peggy Janich
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Salvador A Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| | - David Sassoon
- Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition (ICAN), UMRS 1166 INSERM, University of Pierre and Marie Curie Paris VI, Paris, France
| |
Collapse
|
45
|
Shadrin IY, Khodabukus A, Bursac N. Striated muscle function, regeneration, and repair. Cell Mol Life Sci 2016; 73:4175-4202. [PMID: 27271751 PMCID: PMC5056123 DOI: 10.1007/s00018-016-2285-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022]
Abstract
As the only striated muscle tissues in the body, skeletal and cardiac muscle share numerous structural and functional characteristics, while exhibiting vastly different size and regenerative potential. Healthy skeletal muscle harbors a robust regenerative response that becomes inadequate after large muscle loss or in degenerative pathologies and aging. In contrast, the mammalian heart loses its regenerative capacity shortly after birth, leaving it susceptible to permanent damage by acute injury or chronic disease. In this review, we compare and contrast the physiology and regenerative potential of native skeletal and cardiac muscles, mechanisms underlying striated muscle dysfunction, and bioengineering strategies to treat muscle disorders. We focus on different sources for cellular therapy, biomaterials to augment the endogenous regenerative response, and progress in engineering and application of mature striated muscle tissues in vitro and in vivo. Finally, we discuss the challenges and perspectives in translating muscle bioengineering strategies to clinical practice.
Collapse
Affiliation(s)
- I Y Shadrin
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - A Khodabukus
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - N Bursac
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA.
| |
Collapse
|
46
|
Sojoodi M, Stradiot L, Tanaka K, Heremans Y, Leuckx G, Besson V, Staels W, Van de Casteele M, Marazzi G, Sassoon D, Heimberg H, Bonfanti P. The zinc finger transcription factor PW1/PEG3 restrains murine beta cell cycling. Diabetologia 2016; 59:1474-1479. [PMID: 27130279 PMCID: PMC4901110 DOI: 10.1007/s00125-016-3954-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 03/15/2016] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS Pw1 or paternally-expressed gene 3 (Peg3) encodes a zinc finger transcription factor that is widely expressed during mouse embryonic development and later restricted to multiple somatic stem cell lineages in the adult. The aim of the present study was to define Pw1 expression in the embryonic and adult pancreas and investigate its role in the beta cell cycle in Pw1 wild-type and mutant mice. METHODS We analysed PW1 expression by immunohistochemistry in pancreas of nonpregant and pregnant mice and following injury by partial duct ligation. Its role in the beta cell cycle was studied in vivo using a novel conditional knockout mouse and in vitro by lentivirus-mediated gene knockdown. RESULTS We showed that PW1 is expressed in early pancreatic progenitors at E9.5 but becomes progressively restricted to fully differentiated beta cells as they become established after birth and withdraw from the cell cycle. Notably, PW1 expression declines when beta cells are induced to proliferate and loss of PW1 function activates the beta cell cycle. CONCLUSIONS/INTERPRETATION These results indicate that PW1 is a co-regulator of the beta cell cycle and can thus be considered a novel therapeutic target in diabetes.
Collapse
Affiliation(s)
- Mozhdeh Sojoodi
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Leslie Stradiot
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Karo Tanaka
- Stem Cells and Regenerative Medicine Team, Institute of Cardiology and Nutrition, Inserm UMRS-1166, University Pierre and Marie Curie (Paris VI), Paris, France
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Vanessa Besson
- Stem Cells and Regenerative Medicine Team, Institute of Cardiology and Nutrition, Inserm UMRS-1166, University Pierre and Marie Curie (Paris VI), Paris, France
| | - Willem Staels
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Mark Van de Casteele
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Giovanna Marazzi
- Stem Cells and Regenerative Medicine Team, Institute of Cardiology and Nutrition, Inserm UMRS-1166, University Pierre and Marie Curie (Paris VI), Paris, France
| | - David Sassoon
- Stem Cells and Regenerative Medicine Team, Institute of Cardiology and Nutrition, Inserm UMRS-1166, University Pierre and Marie Curie (Paris VI), Paris, France
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Paola Bonfanti
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK.
- Institute of Immunity and Transplantation, University College London, London, UK.
| |
Collapse
|
47
|
A Novel Mutant Allele of Pw1/Peg3 Does Not Affect Maternal Behavior or Nursing Behavior. PLoS Genet 2016; 12:e1006053. [PMID: 27187722 PMCID: PMC4871489 DOI: 10.1371/journal.pgen.1006053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 04/23/2016] [Indexed: 11/25/2022] Open
Abstract
Parental imprinting is a mammalian-specific form of epigenetic regulation in which one allele of a gene is silenced depending on its parental origin. Parentally imprinted genes have been shown to play a role in growth, metabolism, cancer, and behavior. Although the molecular mechanisms underlying parental imprinting have been largely elucidated, the selective advantage of silencing one allele remains unclear. The mutant phenotype of the imprinted gene, Pw1/Peg3, provides a key example to illustrate the hypothesis on a coadaptation between mother and offspring, in which Pw1/Peg3 is required for a set of essential maternal behaviors, such as nursing, nest building, and postnatal care. We have generated a novel Pw1/Peg3 mutant allele that targets the last exon for the PW1 protein that contains >90% of the coding sequence resulting in a loss of Pw1/Peg3 expression. In contrast to previous reports that have targeted upstream exons, we observe that maternal behavior and lactation are not disrupted upon loss of Pw1/Peg3. Both paternal and homozygous Pw1/Peg3 mutant females nurse and feed their pups properly and no differences are detected in either oxytocin neuron number or oxytocin plasma levels. In addition, suckling capacities are normal in mutant pups. Consistent with previous reports, we observe a reduction of postnatal growth. These results support a general role for Pw1/Peg3 in the regulation of body growth but not maternal care and lactation. Parental genomic imprinting is a mammalian-specific form of epigenetic control that regulates genes differently depending upon whether they are paternally or maternally inherited. The selective advantage of genomic imprinting is poorly understood and has been the subject of numerous theories. In the last several decades, mouse genetic studies have revealed that imprinted genes regulate embryonic and postnatal growth, metabolism, stem cells, neuronal functions, and most notably, behavior. The paternally expressed gene Pw1/Peg3 was one of the first imprinted genes shown to influence maternal behaviors essential for pup survival and growth. Several key studies have demonstrated that Pw1/Peg3 is required for proper nursing and milk ejection by the mother and suckling by the offspring. These previous observations have provided a strong support for the coadaptation theory of imprinting, which proposes that imprinted genes regulate the use of resources between mother and progeny to optimize their survival and future reproductive success. Here we describe that Pw1/Peg3 mutant females exhibit intact maternal behaviors and do not display milk ejection defects. In addition, mutant pups are able to nurse properly.
Collapse
|
48
|
Pierantozzi E, Vezzani B, Badin M, Curina C, Severi FM, Petraglia F, Randazzo D, Rossi D, Sorrentino V. Tissue-Specific Cultured Human Pericytes: Perivascular Cells from Smooth Muscle Tissue Have Restricted Mesodermal Differentiation Ability. Stem Cells Dev 2016; 25:674-86. [PMID: 26956507 DOI: 10.1089/scd.2015.0336] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Microvascular pericytes (PCs) are considered the adult counterpart of the embryonic mesoangioblasts, which represent a multipotent cell population that resides in the dorsal aorta of the developing embryo. Although PCs have been isolated from several adult organs and tissues, it is still controversial whether PCs from different tissues exhibit distinct differentiation potentials. To address this point, we investigated the differentiation potentials of isogenic human cultured PCs isolated from skeletal (sk-hPCs) and smooth muscle tissues (sm-hPCs). We found that both sk-hPCs and sm-hPCs expressed known pericytic markers and did not express endothelial, hematopoietic, and myogenic markers. Both sk-hPCs and sm-hPCs were able to differentiate into smooth muscle cells. In contrast, sk-hPCs, but not sm-hPCs, differentiated in skeletal muscle cells and osteocytes. Given the reported ability of the Notch pathway to regulate skeletal muscle and osteogenic differentiation, sk-hPCs and sm-hPCs were treated with N-[N-(3,5- difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), a known inhibitor of Notch signaling. DAPT treatment, as assessed by histological and molecular analysis, enhanced myogenic differentiation and abolished osteogenic potential of sk-hPCs. In contrast, DAPT treatment did not affect either myogenic or osteogenic differentiation of sm-hPCs. In summary, these results indicate that, despite being isolated from the same anatomical niche, cultured PCs from skeletal muscle and smooth muscle tissues display distinct differentiation abilities.
Collapse
Affiliation(s)
| | - Bianca Vezzani
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Margherita Badin
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Carlo Curina
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Filiberto Maria Severi
- 2 Division of Obstetrics and Gynecology, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Felice Petraglia
- 2 Division of Obstetrics and Gynecology, Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| | - Davide Randazzo
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | - Daniela Rossi
- 1 Molecular Medicine Section, University of Siena , Siena, Italy
| | | |
Collapse
|
49
|
Neill T, Schaefer L, Iozzo RV. Decorin as a multivalent therapeutic agent against cancer. Adv Drug Deliv Rev 2016; 97:174-85. [PMID: 26522384 DOI: 10.1016/j.addr.2015.10.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 12/22/2022]
Abstract
Decorin is a prototypical small leucine-rich proteoglycan that epitomizes the multifunctional nature of this critical gene family. Soluble decorin engages multiple receptor tyrosine kinases within the target-rich environment of the tumor stroma and tumor parenchyma. Upon receptor binding, decorin initiates signaling pathways within endothelial cells downstream of VEGFR2 that ultimately culminate in a Peg3/Beclin 1/LC3-dependent autophagic program. Concomitant with autophagic induction, decorin blunts capillary morphogenesis and endothelial cell migration, thereby significantly compromising tumor angiogenesis. In parallel within the tumor proper, decorin binds multiple RTKs with high affinity, including Met, for a multitude of oncosuppressive functions including growth inhibition, tumor cell mitophagy, and angiostasis. Decorin is also pro-inflammatory by modulating macrophage function and cytokine secretion. Decorin suppresses tumorigenic growth, angiogenesis, and prevents metastatic lesions in a variety of in vitro and in vivo tumor models. Therefore, decorin would be an ideal therapeutic candidate for combating solid malignancies.
Collapse
|
50
|
Dierick F, Héry T, Hoareau-Coudert B, Mougenot N, Monceau V, Claude C, Crisan M, Besson V, Dorfmüller P, Marodon G, Fadel E, Humbert M, Yaniz-Galende E, Hulot JS, Marazzi G, Sassoon D, Soubrier F, Nadaud S. Resident PW1+ Progenitor Cells Participate in Vascular Remodeling During Pulmonary Arterial Hypertension. Circ Res 2016; 118:822-33. [PMID: 26838788 DOI: 10.1161/circresaha.115.307035] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 01/12/2016] [Indexed: 12/20/2022]
Abstract
RATIONALE Pulmonary arterial hypertension is characterized by vascular remodeling and neomuscularization. PW1(+) progenitor cells can differentiate into smooth muscle cells (SMCs) in vitro. OBJECTIVE To determine the role of pulmonary PW1(+) progenitor cells in vascular remodeling characteristic of pulmonary arterial hypertension. METHODS AND RESULTS We investigated their contribution during chronic hypoxia-induced vascular remodeling in Pw1(nLacZ+/-) mouse expressing β-galactosidase in PW1(+) cells and in differentiated cells derived from PW1(+) cells. PW1(+) progenitor cells are present in the perivascular zone in rodent and human control lungs. Using progenitor markers, 3 distinct myogenic PW1(+) cell populations were isolated from the mouse lung of which 2 were significantly increased after 4 days of chronic hypoxia. The number of proliferating pulmonary PW1(+) cells and the proportion of β-gal(+) vascular SMC were increased, indicating a recruitment of PW1(+) cells and their differentiation into vascular SMC during early chronic hypoxia-induced neomuscularization. CXCR4 inhibition using AMD3100 prevented PW1(+) cells differentiation into SMC but did not inhibit their proliferation. Bone marrow transplantation experiments showed that the newly formed β-gal(+) SMC were not derived from circulating bone marrow-derived PW1(+) progenitor cells, confirming a resident origin of the recruited PW1(+) cells. The number of pulmonary PW1(+) cells was also increased in rats after monocrotaline injection. In lung from pulmonary arterial hypertension patients, PW1-expressing cells were observed in large numbers in remodeled vascular structures. CONCLUSIONS These results demonstrate the existence of a novel population of resident SMC progenitor cells expressing PW1 and participating in pulmonary hypertension-associated vascular remodeling.
Collapse
Affiliation(s)
- France Dierick
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Tiphaine Héry
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Bénédicte Hoareau-Coudert
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Nathalie Mougenot
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Virginie Monceau
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Caroline Claude
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Mihaela Crisan
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Vanessa Besson
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Peter Dorfmüller
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Gilles Marodon
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Elie Fadel
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Marc Humbert
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Elisa Yaniz-Galende
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Jean-Sébastien Hulot
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Giovanna Marazzi
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - David Sassoon
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Florent Soubrier
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.)
| | - Sophie Nadaud
- From the INSERM, Institute of Cardiometabolism and Nutrition, UMR_S 1166-ICAN (F.D., T.H., V.M., C.C., V.B., E.Y.-G., J.-S.H., G.M., D.S., F.S., S.N.), UMS-030 CyPS, Paris, France (B.H.-C.), PECMV UMS28 (N.M.), INSERM, CNRS, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI), U1135, ERL 8255 (G.M.), Sorbonne Universités, UPMC Univ Paris 06, Paris, France; Erasmus MC Stem Cell Institute, Rotterdam, The Netherlands (M.C.); Univ Paris-Sud, Université Paris Saclay, INSERM UMR-S 999, Labex LERMIT, Le Plessis-Robinson, Paris, France (P.D., E.F., M.H.); Service d'Anatomie Pathologique, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, Paris, France (P.D.); Service de Chirurgie Thoracique et Vasculaire, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (E.F.); Univ Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, Paris, France (M.H.); and Assistance Publique Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Hôpital Bicêtre, Le Kremlin Bicêtre, France (M.H.).
| |
Collapse
|