1
|
Florczyk-Soluch U, Polak K, Jelinkova S, Bronisz-Budzyńska I, Sabo R, Bolisetty S, Agarwal A, Werner E, Józkowicz A, Stępniewski J, Szade K, Dulak J. Targeted expression of heme oxygenase-1 in satellite cells improves skeletal muscle pathology in dystrophic mice. Skelet Muscle 2024; 14:13. [PMID: 38867250 PMCID: PMC11167827 DOI: 10.1186/s13395-024-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/03/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.
Collapse
MESH Headings
- Animals
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Mice, Inbred mdx
- Mice, Transgenic
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mice
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Male
- Mice, Inbred C57BL
- Physical Conditioning, Animal
- Membrane Proteins
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Sarka Jelinkova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Reece Sabo
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ewa Werner
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Jacek Stępniewski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Laboratory of Stem Cells Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
2
|
Dar A, Li A, Petrigliano FA. Lineage tracing reveals a novel PDGFRβ + satellite cell subset that contributes to myo-regeneration of chronically injured rotator cuff muscle. Sci Rep 2024; 14:9668. [PMID: 38671006 PMCID: PMC11053018 DOI: 10.1038/s41598-024-58926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Massive rotator cuff (RC) tendon tears are associated with progressive fibro-adipogenesis and muscle atrophy that altogether cause shoulder muscle wasting. Platelet derived growth factor β (PDGFRβ) lineage cells, that co-express PDGFRα have previously been shown to directly contribute to scar formation and fat accumulation in a mouse model of irreversible tendon and nerve transection (TTDN). Conversely, PDGFRβ+ lineage cells have also been shown to be myogenic in cultures and in other models of skeletal muscle injury. We therefore hypothesized that PDGFRβ demarcates two distinct RC residing subpopulations, fibro-adipogenic and myogenic, and aimed to elucidate the identity of the PDGFRβ myogenic precursors and evaluate their contribution, if any, to RC myo-regeneration. Lineage tracing revealed increasing contribution of PDGFRβ+ myo-progenitors to the formation of GFP+ myofibers, which were the most abundant myofiber type in regenerated muscle at 2 weeks post-TTDN. Muscle regeneration preceded muscle atrophy and both advanced from the lateral site of tendon transection to the farthest medial region. GFP+/PDGFRβ+Sca-1-lin-CXCR4+Integrin-β1+ marked a novel subset of satellite cells with confirmed myogenic properties. Further studies are warranted to identify the existence of PDGFRβ+ satellite cells in human and other mouse muscles and to define their myo-regenerative potential following acute and chronic muscle injury.
Collapse
Affiliation(s)
- Ayelet Dar
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Angela Li
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
4
|
Liu D, Xu C, Liu Y, Ouyang W, Lin S, Xu A, Zhang Y, Xie Y, Huang Q, Zhao W, Chen Z, Wang L, Chen S, Huang J, Wu ZB, Sun X. A systematic survey of LU domain-containing proteins reveals a novel human gene, LY6A, which encodes the candidate ortholog of mouse Ly-6A/Sca-1 and is aberrantly expressed in pituitary tumors. Front Med 2023; 17:458-475. [PMID: 36928550 DOI: 10.1007/s11684-022-0968-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 10/12/2022] [Indexed: 03/18/2023]
Abstract
The Ly-6 and uPAR (LU) domain-containing proteins represent a large family of cell-surface markers. In particular, mouse Ly-6A/Sca-1 is a widely used marker for various stem cells; however, its human ortholog is missing. In this study, based on a systematic survey and comparative genomic study of mouse and human LU domain-containing proteins, we identified a previously unannotated human gene encoding the candidate ortholog of mouse Ly-6A/Sca-1. This gene, hereby named LY6A, reversely overlaps with a lncRNA gene in the majority of exonic sequences. We found that LY6A is aberrantly expressed in pituitary tumors, but not in normal pituitary tissues, and may contribute to tumorigenesis. Similar to mouse Ly-6A/Sca-1, human LY6A is also upregulated by interferon, suggesting a conserved transcriptional regulatory mechanism between humans and mice. We cloned the full-length LY6A cDNA, whose encoded protein sequence, domain architecture, and exon-intron structures are all well conserved with mouse Ly-6A/Sca-1. Ectopic expression of the LY6A protein in cells demonstrates that it acts the same as mouse Ly-6A/Sca-1 in their processing and glycosylphosphatidylinositol anchoring to the cell membrane. Collectively, these studies unveil a novel human gene encoding a candidate biomarker and provide an interesting model gene for studying gene regulatory and evolutionary mechanisms.
Collapse
Affiliation(s)
- Dan Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chunhui Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanting Liu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wen Ouyang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shaojian Lin
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Aining Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuanliang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qiuhua Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weili Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jinyan Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Biomedical Big Data Center, First Affiliated Hospital, Zhejiang University School of Medicine, and Cancer Center, Zhejiang University, Hangzhou, 310000, China.
| | - Zhe Bao Wu
- Department of Neurosurgery, Center of Pituitary Tumor, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xiaojian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
5
|
Production of Stem Cell Antigen 1 Sca-1/Ly-6A/E by Freshly Isolated NK Cells Cultured with Relevant Cytokines. Bull Exp Biol Med 2022; 174:62-65. [PMID: 36437320 DOI: 10.1007/s10517-022-05649-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 11/29/2022]
Abstract
Activated NK cells in appropriate conditions are known to express stem cell antigen 1 (Sca-1/Ly-6A/E). To investigate its production, NK cells isolated from mouse spleens were incubated ex vivo in the presence of different combinations of cytokines (IL-12, IL-15, IL-18, and IFNγ). Expression of Sca-1 was found to be considerably higher in NK cells incubated in the presence of IL-18, IL-15, and IL-12 than in those treated with IL-15 and IL-18 only. To test the hypothesis that the effect of IL-12 was due to stimulation of IFNγ production, we replaced IL-12 with IFNγ in some samples and added specific anti-IFNγ antibody to some samples cultured with IL-15/IL-18+IL-12. In the subpopulations incubated in the presence of IL-15/IL-18 with added IFNγ instead of IL-12, the expression of Sca-1 was not increased. By contrast, in samples treated with IL-15/IL-18+IL-12 and anti-IFNγ antibody, the expression of Sca-1 was activated to a similar extent as in those stimulated by IL-15/IL-18+IL-12 combination without the antibody. The obtained data suggest that IL-12 activates the production of Sca-1 by NK cells through an IFNγ-independent mechanism.
Collapse
|
6
|
LaCombe R, Cecchini A, Seibert M, Cornelison DDW. EphA1 receptor tyrosine kinase is localized to the nucleus in rhabdomyosarcoma from multiple species. Biol Open 2022; 11:bio059352. [PMID: 36214254 PMCID: PMC9581518 DOI: 10.1242/bio.059352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022] Open
Abstract
While the typical role of receptor tyrosine kinases is to receive and transmit signals at the cell surface, in some cellular contexts (particularly transformed cells) they may also act as nuclear proteins. Aberrant nuclear localization of receptor tyrosine kinases associated with transformation often enhances the transformed phenotype (i.e. nuclear ErbBs promote tumor progression in breast cancer). Rhabdomyosarcoma (RMS), the most common soft tissue tumor in children, develops to resemble immature skeletal muscle and has been proposed to derive from muscle stem/progenitor cells (satellite cells). It is an aggressive cancer with a 5-year survival rate of 33% if it has metastasized. Eph receptor tyrosine kinases have been implicated in the development and progression of many other tumor types, but there are only two published studies of Ephs localizing to the nucleus of any cell type and to date no nuclear RTKs have been identified in RMS. In a screen for protein expression of Ephs in canine RMS primary tumors as well as mouse and human RMS cell lines, we noted strong expression of EphA1 in the nucleus of interphase cells in tumors from all three species. This localization pattern changes in dividing cells, with EphA1 localizing to the nucleus or the cytoplasm depending on the phase of the cell cycle. These data represent the first case of a nuclear RTK in RMS, and the first time that EphA1 has been detected in the nucleus of any cell type.
Collapse
Affiliation(s)
- Ronnie LaCombe
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Alessandra Cecchini
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Morgan Seibert
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
| | - DDW Cornelison
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
7
|
TNF-α Suppresses Apelin Receptor Expression in Mouse Quadriceps Femoris-Derived Cells. Curr Issues Mol Biol 2022; 44:3146-3155. [PMID: 35877441 PMCID: PMC9315797 DOI: 10.3390/cimb44070217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Expression of the apelin receptor, APJ, in skeletal muscle (SM) is known to decrease with age, but the underlying mechanism remains unclear. Increased tumor necrosis factor (TNF)-α levels are observed in SM with age and are associated with muscle atrophy. To investigate the possible interconnection between TNF-α elevation and APJ reduction with aging, we investigated the effect of TNF-α on APJ expression in cells derived from the quadriceps femoris of C57BL/6J mice. Expression of Tnfa and Apj in the quadriceps femoris was compared between 4- (young) and 24-month-old (old) C57BL/6J mice (n = 10 each) using qPCR. Additionally, APJ-positive cells and TNF-α protein were analyzed by flow cytometry and Western blotting, respectively. Further, quadricep-derived cells were exposed to 0 (control) or 25 ng/mL TNF-α, and the effect on Apj expression was examined by qRT-PCR. Apj expression and the ratio of APJ-positive cells among quadricep cells were significantly lower in old compared to young mice. In contrast, levels of Tnfa mRNA and TNF-α protein were significantly elevated in old compared to young mice. Exposing young and old derived quadricep cells to TNF-α for 8 and 24 h caused Apj levels to significantly decrease. TNF-α suppresses APJ expression in muscle cells in vitro. The increase in TNF-α observed in SM with age may induce a decrease in APJ expression.
Collapse
|
8
|
Marín-Llera JC, Lorda-Diez CI, Hurle JM, Chimal-Monroy J. SCA-1/Ly6A Mesodermal Skeletal Progenitor Subpopulations Reveal Differential Commitment of Early Limb Bud Cells. Front Cell Dev Biol 2021; 9:656999. [PMID: 34336823 PMCID: PMC8322737 DOI: 10.3389/fcell.2021.656999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/21/2021] [Indexed: 01/14/2023] Open
Abstract
At early developmental stages, limb bud mesodermal undifferentiated cells are morphologically indistinguishable. Although the identification of several mesodermal skeletal progenitor cell populations has been recognized, in advanced stages of limb development here we identified and characterized the differentiation hierarchy of two new early limb bud subpopulations of skeletal progenitors defined by the differential expression of the SCA-1 marker. Based on tissue localization of the mesenchymal stromal cell-associated markers (MSC-am) CD29, Sca-1, CD44, CD105, CD90, and CD73, we identified, by multiparametric analysis, the presence of cell subpopulations in the limb bud capable of responding to inductive signals differentially, namely, sSca+ and sSca– cells. In concordance with its gene expression profile, cell cultures of the sSca+ subpopulation showed higher osteogenic but lower chondrogenic capacity than those of sSca–. Interestingly, under high-density conditions, fibroblast-like cells in the sSca+ subpopulation were abundant. Gain-of-function employing micromass cultures and the recombinant limb assay showed that SCA-1 expression promoted tenogenic differentiation, whereas chondrogenesis is delayed. This model represents a system to determine cell differentiation and morphogenesis of different cell subpopulations in similar conditions like in vivo. Our results suggest that the limb bud is composed of a heterogeneous population of progenitors that respond differently to local differentiation inductive signals in the early stages of development, where SCA-1 expression may play a permissive role during cell fate.
Collapse
Affiliation(s)
- Jessica Cristina Marín-Llera
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, México
| | - Carlos Ignacio Lorda-Diez
- Departamento de Anatomía y Biología Celular and IDIVAL, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Juan Mario Hurle
- Departamento de Anatomía y Biología Celular and IDIVAL, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Jesús Chimal-Monroy
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, México
| |
Collapse
|
9
|
Kapoor S, Subba P, Shenoy P S, Bose B. Sca1 + Progenitor Cells (Ex vivo) Exhibits Differential Proteomic Signatures From the Culture Adapted Sca1 + Cells (In vitro), Both Isolated From Murine Skeletal Muscle Tissue. Stem Cell Rev Rep 2021; 17:1754-1767. [PMID: 33742350 DOI: 10.1007/s12015-021-10134-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 10/21/2022]
Abstract
Stem cell antigen-1 (Sca-1) is a glycosyl-phosphatidylinositol-anchored membrane protein that is expressed in a sub-population of muscle stem and progenitor cell types. Reportedly, Sca-1 regulates the myogenic property of myoblasts and Sca-1-/- mice exhibited defective muscle regeneration. Although the role of Sca-1 in muscle development and maintenance is well-acknowledged, molecular composition of muscle derived Sca-1+ cells is not characterized. Here, we applied a high-resolution mass spectrometry-based workflow to characterize the proteomic landscape of mouse hindlimb skeletal muscle derived Sca-1+ cells. Furthermore, we characterized the impact of the cellular microenvironments on the proteomes of Sca-1+ cells. The proteome component of freshly isolated Sca-1+ cells (ex vivo) was compared with that of Sca-1+ cells expanded in cell culture (in vitro). The analysis revealed significant differences in the protein abundances in the two conditions reflective of their functional variations. The identified proteins were enriched in various biological pathways. Notably, we identified proteins related to myotube differentiation, myotube cell development and myoblast fusion. We also identified a panel of cell surface marker proteins that can be leveraged in future to enrich Sca-1+ cells using combinatorial strategies. Comparative analysis implicated the activation of various pathways leading to increased protein synthesis under in vitro condition. We report here the most comprehensive proteome map of Sca-1+ cells that provides insights into the molecular networks operative in Sca-1+ cells. Importantly, through our work we generated the proteomic blueprint of protein abundances significantly altered in Sca-1+ cells under ex vivo and in vitro conditions. The curated data can also be visualized at https://yenepoya.res.in/database/Sca-1-Proteomics .
Collapse
Affiliation(s)
- Saketh Kapoor
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Pratigya Subba
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
10
|
Christensen R, Gunnarsson AP, Jensen UB. The role of stem cell antigen-1/Lymphocyte antigen 6A-2/6E-1 knock out in murine epidermis. Stem Cell Res 2020; 49:102047. [PMID: 33157392 DOI: 10.1016/j.scr.2020.102047] [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: 07/07/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 11/27/2022] Open
Abstract
Stem Cell Antigen-1 (SCA-1) is a central positive marker for isolating stem cells in several tissues in the mouse. However, for the epidermis, this appears to be the opposite since lack of SCA-1 has been shown to identify keratinocyte populations with progenitor characteristics. This study investigates the effect of SCA-1 knockout in murine keratinocytes. We compared Sca-1EGFP/EGFP knockout and wildtype mice with respect to the three-dimensional morphology of the epidermis, performed functional assays, and generated gene expression profiles on FACS sorted cells. There were no morphological abnormalities on skin, fur, or hair follicles in transgenic knockout mice compared to wild type mice. SCA-1 knockout keratinocytes showed significantly reduced colony-forming efficiency, colony size and proliferation rate in vitro, however, SCA-1 knockout did not alter wound healing efficiency or keratinocyte proliferation rate in vivo. Moreover, gene expression profiling shows that the effect from knockout of SCA-1 in keratinocytes is dissimilar from what has been observed in other tissues. Additionally, tumor assay indicated that SCA-1 knockout decreases the number of formed papillomas. The results indicate a more complex role for SCA-1, which might differ between epidermal keratinocytes during homeostasis and activated conditions.
Collapse
Affiliation(s)
- Rikke Christensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| | - Anders Patrik Gunnarsson
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Brendstrupgaardsvej 21C, 8200 Aarhus N, Denmark.
| |
Collapse
|
11
|
IL-12 regulates the expansion, phenotype, and function of murine NK cells activated by IL-15 and IL-18. Cancer Immunol Immunother 2020; 69:1699-1712. [PMID: 32333080 DOI: 10.1007/s00262-020-02553-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 03/19/2020] [Indexed: 12/12/2022]
Abstract
NK cells, which are composed of phenotypically and functionally heterogeneous subpopulations, play critical roles in immunity against cancer. The mechanism of generation of distinct subsets such as the effector and regulatory subtypes is unclear. Here, we show that this process comprises several steps, including generation of proliferating, highly cytotoxic cells activated by IL-15/IL-18 and differentiation into distinct cell populations induced with IL-12. Freshly prepared murine splenic NK cells expressed IL-15Rs and IL-18Rs and rapidly began to proliferate following stimulation with IL-15/IL-18. The proliferating NK cells highly expressed various activation markers such as B220, CD49b (DX5), lysosome-associated membrane glycoprotein 1 (LAMP-1), DNAX accessory molecule 1, perforin, and granzyme B and showed reduced expression of natural killer cell p46-related protein (NKp46) and IL-18Rα. These cells exerted strong cytotoxicity against YAC-1 cells, but did not secrete cytokines. IL-12 rapidly activated STAT4 in these cells, induced IFN-γ production, and then upregulated p21 and p27, leading to withdrawal from the cell cycle. In parallel, IL-12-stimulated cells gradually reduced cytotoxicity, decreased expression of activation markers, and instead increased expression of Sca-1, CD25, CD49a, and NKp46. Some IL-15/IL-18-induced cells strongly expressed PD-1, whereas NK cells induced with IL-15/IL-18 and IL-12 expressed high levels of T cell immunoglobulin mucin-3, LAG-3, and natural killer group 2 A. Furthermore, these cells spontaneously secreted IL-10 and TGF-β following prolonged incubation. Thus, IL-12 regulates expansion of NK cells activated with IL-15/IL-18, influences the population size of highly cytotoxic cells, and induces differentiation to unique cells sharing some phenotypes of ILCs.
Collapse
|
12
|
A Three-Dimensional Culture Model of Reversibly Quiescent Myogenic Cells. Stem Cells Int 2019; 2019:7548160. [PMID: 31827532 PMCID: PMC6885280 DOI: 10.1155/2019/7548160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/24/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Satellite cells (SC) are the stem cells of skeletal muscles. They are quiescent in adult animals but resume proliferation to allow muscle hypertrophy or regeneration after injury. The mechanisms balancing quiescence, self-renewal, and differentiation of SC are difficult to analyze in vivo owing to their complexity and in vitro because the staminal character of SC is lost when they are removed from the niche and is not adequately reproduced in the culture models currently available. To overcome these difficulties, we set up a culture model of the myogenic C2C12 cell line in suspension. When C2C12 cells are cultured in suspension, they enter a state of quiescence and form three-dimensional aggregates (myospheres) that produce the extracellular matrix and express markers of quiescent SC. In the initial phase of culture, a portion of the cells fuses in syncytia and abandons the myospheres. The remaining cells are mononucleated and quiescent but resume proliferation and differentiation when plated in a monolayer. The notch pathway controls the quiescent state of the cells as shown by the fact that its inhibition leads to the resumption of differentiation. Within this context, notch3 appears to play a central role in the activity of this pathway since the expression of notch1 declines soon after aggregation. In summary, the culture model of C2C12 in suspension may be used to study the cellular interactions of muscle stem cells and the pathways controlling SC quiescence entrance and maintenance.
Collapse
|
13
|
Role of Mutant TBP in Regulation of Myogenesis on Muscle Satellite Cells. Curr Med Sci 2019; 39:734-740. [PMID: 31612390 DOI: 10.1007/s11596-019-2099-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/24/2019] [Indexed: 12/15/2022]
Abstract
In polyglutamine (PolyQ) diseases, mutant proteins cause not only neurological problems but also peripheral tissue abnormalities. Among all systemic damages, skeletal muscle dystrophy is the severest. Previously by studying knock-in (KI) mouse models of spinal cerebellar ataxia 17 (SCA17), it was found that mutant TATA box binding protein (TBP) decreases its interaction with myogenic differentiation antigen, thus reducing the expression of skeletal muscle structural proteins and resulting in muscle degeneration. In this paper, the role of mutant TBP in myogenesis was investigated. Single myofibers were isolated from tibialis anterior muscles of wild type (WT) and SCA17KI mice. The 1TBP18 staining confirmed the expression of mutant TBP in muscle satellite cells in SCA17KI mice. In the BaCl2-induced TA muscle injury, H&E cross-section staining showed no significant change in myofibril size before and after BaCl2 treatment, and there was no significant difference in centralized nuclei between WT and SCA17KI mice, suggesting that mutant TBP had no significant effect on muscle regeneration. In the cultured primary myoblasts from WT and SCA17KI mice in vitro, representative BrdU immunostaining showed no significant difference in proliferation of muscle satellite cells. The primary myoblasts were then induced to differentiate and immunostained for eMyHC, and the staining showed there was no significant difference in differentiation of primary myoblasts between WT and SCA1KI mice. Our findings confirmed that mutant TBP had no significant effect on myogenesis.
Collapse
|
14
|
Marinkovic M, Fuoco C, Sacco F, Cerquone Perpetuini A, Giuliani G, Micarelli E, Pavlidou T, Petrilli LL, Reggio A, Riccio F, Spada F, Vumbaca S, Zuccotti A, Castagnoli L, Mann M, Gargioli C, Cesareni G. Fibro-adipogenic progenitors of dystrophic mice are insensitive to NOTCH regulation of adipogenesis. Life Sci Alliance 2019; 2:e201900437. [PMID: 31239312 PMCID: PMC6599969 DOI: 10.26508/lsa.201900437] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022] Open
Abstract
Fibro-adipogenic progenitors (FAPs) promote satellite cell differentiation in adult skeletal muscle regeneration. However, in pathological conditions, FAPs are responsible for fibrosis and fatty infiltrations. Here we show that the NOTCH pathway negatively modulates FAP differentiation both in vitro and in vivo. However, FAPs isolated from young dystrophin-deficient mdx mice are insensitive to this control mechanism. An unbiased mass spectrometry-based proteomic analysis of FAPs from muscles of wild-type and mdx mice suggested that the synergistic cooperation between NOTCH and inflammatory signals controls FAP differentiation. Remarkably, we demonstrated that factors released by hematopoietic cells restore the sensitivity to NOTCH adipogenic inhibition in mdx FAPs. These results offer a basis for rationalizing pathological ectopic fat infiltrations in skeletal muscle and may suggest new therapeutic strategies to mitigate the detrimental effects of fat depositions in muscles of dystrophic patients.
Collapse
Affiliation(s)
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Giulio Giuliani
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Alessio Reggio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Filomena Spada
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Simone Vumbaca
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
15
|
Pencheva-Demireva M, El-Darawish Y, Kavaldzhieva K, Mladenov N, Lazarov V, Dimitrova-Dikanarova D, Markova T, Nikolov R, Okamura H. Upregulation of Natural Killer Cells Proliferation by Cytokine Stimulation. Monoclon Antib Immunodiagn Immunother 2019; 38:85-88. [PMID: 31009333 DOI: 10.1089/mab.2019.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Natural killer (NK) cells can discriminate between normal and cancer cells and are known to directly recognize and kill malignant cells or induce apoptosis. Thus, activation of NK cells is considered as a promising strategy for cancer treatment. However, clinical application has been somewhat limited because of difficulties in the preparation of sufficient number of highly cytotoxic/activated NK cells in vitro. We used cytokine stimulation to provide a suitable environment (activating receptor-ligand interactions) for the expansion of NK cells. This method potently expanded NK cells, and the final product was composed of highly proliferating NK cells. The expanded NK cells showed significant upregulation of various activation receptors such as CD69 and NKG2D. The latter is a particularly important receptor for triggering NK cell responses toward tumor cells.
Collapse
Affiliation(s)
- Magdalena Pencheva-Demireva
- 1 Department of Medical Biology, Medical Faculty, Medical University, Sofia, Bulgaria.,2 Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yosif El-Darawish
- 2 Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| | - Katerina Kavaldzhieva
- 1 Department of Medical Biology, Medical Faculty, Medical University, Sofia, Bulgaria
| | - Nikola Mladenov
- 1 Department of Medical Biology, Medical Faculty, Medical University, Sofia, Bulgaria
| | - Vladislav Lazarov
- 1 Department of Medical Biology, Medical Faculty, Medical University, Sofia, Bulgaria
| | | | - Tzvetanka Markova
- 3 Department of Pharmacology, Medical Faculty, Medical University, Sofia, Bulgaria
| | - Rumen Nikolov
- 3 Department of Pharmacology, Medical Faculty, Medical University, Sofia, Bulgaria
| | - Haruki Okamura
- 2 Laboratory of Tumor Immunology and Cell Therapy, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
16
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
17
|
Constitutive transgene expression of Stem Cell Antigen-1 in the hair follicle alters the sensitivity to tumor formation and progression. Stem Cell Res 2017; 23:109-118. [DOI: 10.1016/j.scr.2017.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/06/2017] [Indexed: 02/05/2023] Open
|
18
|
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
|
19
|
Penton CM, Badarinarayana V, Prisco J, Powers E, Pincus M, Allen RE, August PR. Laminin 521 maintains differentiation potential of mouse and human satellite cell-derived myoblasts during long-term culture expansion. Skelet Muscle 2016; 6:44. [PMID: 27964750 PMCID: PMC5154152 DOI: 10.1186/s13395-016-0116-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/01/2016] [Indexed: 12/03/2022] Open
Abstract
Background Large-scale expansion of myogenic progenitors is necessary to support the development of high-throughput cellular assays in vitro and to advance genetic engineering approaches necessary to develop cellular therapies for rare muscle diseases. However, optimization has not been performed in order to maintain the differentiation capacity of myogenic cells undergoing long-term cell culture. Multiple extracellular matrices have been utilized for myogenic cell studies, but it remains unclear how different matrices influence long-term myogenic activity in culture. To address this challenge, we have evaluated multiple extracellular matrices in myogenic studies over long-term expansion. Methods We evaluated the consequence of propagating mouse and human myogenic stem cell progenitors on various extracellular matrices to determine if they could enhance long-term myogenic potential. For the first time reported, we comprehensively examine the effect of physiologically relevant laminins, laminin 211 and laminin 521, compared to traditionally utilized ECMs (e.g., laminin 111, gelatin, and Matrigel) to assess their capacity to preserve myogenic differentiation potential. Results Laminin 521 supported increased proliferation in early phases of expansion and was the only substrate facilitating high-level fusion following eight passages in mouse myoblast cell cultures. In human myoblast cell cultures, laminin 521 supported increased proliferation during expansion and superior differentiation with myotube hypertrophy. Counterintuitively however, laminin 211, the native laminin isoform in resting skeletal muscle, resulted in low proliferation and poor differentiation in mouse and human cultures. Matrigel performed excellent in short-term mouse studies but showed high amounts of variability following long-term expansion. Conclusions These results demonstrate laminin 521 is a superior substrate for both short-term and long-term myogenic cell culture applications compared to other commonly utilized substrates. Since Matrigel cannot be used for clinical applications, we propose that laminin 521 could possibly be employed in the future to provide myoblasts for cellular therapy directed clinical studies. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0116-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher M Penton
- Discovery Biology, Tucson Innovation Center, Icagen, Oro Valley, AZ, 85755, USA.
| | | | - Joy Prisco
- Discovery Biology, Tucson Innovation Center, Sanofi, Oro Valley, AZ, 85755, USA
| | - Elaine Powers
- Discovery Biology, Tucson Innovation Center, Sanofi, Oro Valley, AZ, 85755, USA
| | - Mark Pincus
- Discovery Biology, Tucson Innovation Center, Icagen, Oro Valley, AZ, 85755, USA
| | - Ronald E Allen
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, 85721, USA.
| | - Paul R August
- Discovery Biology, Tucson Innovation Center, Icagen, Oro Valley, AZ, 85755, USA.
| |
Collapse
|
20
|
Pisconti A, Banks GB, Babaeijandaghi F, Betta ND, Rossi FMV, Chamberlain JS, Olwin BB. Loss of niche-satellite cell interactions in syndecan-3 null mice alters muscle progenitor cell homeostasis improving muscle regeneration. Skelet Muscle 2016; 6:34. [PMID: 27757223 PMCID: PMC5064903 DOI: 10.1186/s13395-016-0104-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 08/26/2016] [Indexed: 02/04/2023] Open
Abstract
Background The skeletal muscle stem cell niche provides an environment that maintains quiescent satellite cells, required for skeletal muscle homeostasis and regeneration. Syndecan-3, a transmembrane proteoglycan expressed in satellite cells, supports communication with the niche, providing cell interactions and signals to maintain quiescent satellite cells. Results Syndecan-3 ablation unexpectedly improves regeneration in repeatedly injured muscle and in dystrophic mice, accompanied by the persistence of sublaminar and interstitial, proliferating myoblasts. Additionally, muscle aging is improved in syndecan-3 null mice. Since syndecan-3 null myofiber-associated satellite cells downregulate Pax7 and migrate away from the niche more readily than wild type cells, syxndecan-3 appears to regulate satellite cell homeostasis and satellite cell homing to the niche. Conclusions Manipulating syndecan-3 provides a promising target for development of therapies to enhance muscle regeneration in muscular dystrophies and in aged muscle. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0104-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Addolorata Pisconti
- Department of Cellular, Molecular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309 USA ; Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB UK
| | - Glen B Banks
- Department of Neurology, University of Washington, Mail Stop 357720, Seattle, WA 98195 USA
| | | | - Nicole Dalla Betta
- Department of Cellular, Molecular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309 USA
| | - Fabio M V Rossi
- The Biomedical Research Centre, UBC, Vancouver, BC V6T 1Z Canada
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington, Mail Stop 357720, Seattle, WA 98195 USA
| | - Bradley B Olwin
- Department of Cellular, Molecular and Developmental Biology, University of Colorado at Boulder, Boulder, CO 80309 USA
| |
Collapse
|
21
|
Smeriglio P, Alonso-Martin S, Masciarelli S, Madaro L, Iosue I, Marrocco V, Relaix F, Fazi F, Marazzi G, Sassoon DA, Bouché M. Phosphotyrosine phosphatase inhibitor bisperoxovanadium endows myogenic cells with enhanced muscle stem cell functions via epigenetic modulation of Sca-1 and Pw1 promoters. FASEB J 2015; 30:1404-15. [PMID: 26672000 DOI: 10.1096/fj.15-275420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023]
Abstract
Understanding the regulation of the stem cell fate is fundamental for designing novel regenerative medicine strategies. Previous studies have suggested that pharmacological treatments with small molecules provide a robust and reversible regulation of the stem cell program. Previously, we showed that treatment with a vanadium compound influences muscle cell fatein vitro In this study, we demonstrate that treatment with the phosphotyrosine phosphatase inhibitor bisperoxovanadium (BpV) drives primary muscle cells to a poised stem cell stage, with enhanced function in muscle regenerationin vivofollowing transplantation into injured muscles. Importantly, BpV-treated cells displayed increased self-renewal potentialin vivoand replenished the niche in both satellite and interstitial cell compartments. Moreover, we found that BpV treatment induces specific activating chromatin modifications at the promoter regions of genes associated with stem cell fate, includingSca-1andPw1 Thus, our findings indicate that BpV resets the cell fate program by specific epigenetic regulations, such that the committed myogenic cell fate is redirected to an earlier progenitor cell fate stage, which leads to an enhanced regenerative stem cell potential.-Smeriglio, P., Alonso-Martin, S., Masciarelli, S., Madaro, L., Iosue, I., Marrocco, V., Relaix, F., Fazi, F., Marazzi, G., Sassoon, D. A., Bouché, M. Phosphotyrosine phosphatase inhibitor bisperoxovanadium endows myogenic cells with enhanced muscle stem cell functionsviaepigenetic modulation of Sca-1 and Pw1 promoters.
Collapse
Affiliation(s)
- Piera Smeriglio
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Sonia Alonso-Martin
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Silvia Masciarelli
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Luca Madaro
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Ilaria Iosue
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Valeria Marrocco
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Frédéric Relaix
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Francesco Fazi
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Giovanna Marazzi
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - David A Sassoon
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Marina Bouché
- *Stem Cells and Regenerative Medicine, Institute of Cardiometabolism and Nutrition Unité Mixte de Recherche en Santé 1166 INSERM/Sorbonne University (Pierre and Marie Curie University, Paris VI), Paris, France; Department of Anatomy, Histology, Forensic Medicine, and Orthopedics, Unit of Histology, Sapienza University of Rome, Rome, Italy; INSERM Unité 955 Institut Mondor de Recherche Biomédicale, Creteil, France; Université Paris-Est Créteil, Faculty of Medicine, Creteil, France; Sorbonne Universités, Pierre and Marie Curie University, Paris VI, INSERM Unité Mixte de Recherche en Santé 974, Centre National de la Recherche Scientifique FRE3617, Center for Research in Myology, Paris, France; Etablissement Français du Sang, Creteil, France; and Université Paris Est, Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| |
Collapse
|
22
|
Pelosi L, Berardinelli MG, Forcina L, Spelta E, Rizzuto E, Nicoletti C, Camilli C, Testa E, Catizone A, De Benedetti F, Musarò A. Increased levels of interleukin-6 exacerbate the dystrophic phenotype in mdx mice. Hum Mol Genet 2015; 24:6041-53. [PMID: 26251044 PMCID: PMC4599671 DOI: 10.1093/hmg/ddv323] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/03/2015] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive lethal muscle degeneration and chronic inflammatory response. The mdx mouse strain has served as the animal model for human DMD. However, while DMD patients undergo extensive necrosis, the affected muscles of adult mdx mice rapidly regenerates and regains structural and functional integrity. The basis for the mild effects observed in mice compared with the lethal consequences in humans remains unknown. In this study, we provide evidence that interleukin-6 (IL-6) is causally linked to the pathogenesis of muscular dystrophy. We report that forced expression of IL-6, in the adult mdx mice, recapitulates the severe phenotypic characteristics of DMD in humans. Increased levels of IL-6 exacerbate the dystrophic muscle phenotype, sustaining inflammatory response and repeated cycles of muscle degeneration and regeneration, leading to exhaustion of satellite cells. The mdx/IL6 mouse closely approximates the human disease and more faithfully recapitulates the disease progression in humans. This study promises to significantly advance our understanding of the pathogenic mechanisms that lead to DMD.
Collapse
Affiliation(s)
- Laura Pelosi
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | | | - Laura Forcina
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Elisa Spelta
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Rome 00184, Italy
| | - Carmine Nicoletti
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Carlotta Camilli
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Erika Testa
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Angela Catizone
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome 00161, Italy
| | | | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy
| |
Collapse
|
23
|
Camarata TD, Weaver GC, Vasilyev A, Arnaout MA. Negative Regulation of TGFβ Signaling by Stem Cell Antigen-1 Protects against Ischemic Acute Kidney Injury. PLoS One 2015; 10:e0129561. [PMID: 26053644 PMCID: PMC4460127 DOI: 10.1371/journal.pone.0129561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/10/2015] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury, often caused by an ischemic insult, is associated with significant short-term morbidity and mortality, and increased risk of chronic kidney disease. The factors affecting the renal response to injury following ischemia and reperfusion remain to be clarified. We found that the Stem cell antigen-1 (Sca-1), commonly used as a stem cell marker, is heavily expressed in renal tubules of the adult mouse kidney. We evaluated its potential role in the kidney using Sca-1 knockout mice submitted to acute ischemia reperfusion injury (IRI), as well as cultured renal proximal tubular cells in which Sca-1 was stably silenced with shRNA. IRI induced more severe injury in Sca-1 null kidneys, as assessed by increased expression of Kim-1 and Ngal, rise in serum creatinine, abnormal pathology, and increased apoptosis of tubular epithelium, and persistent significant renal injury at day 7 post IRI, when recovery of renal function in control animals was nearly complete. Serum creatinine, Kim-1 and Ngal were slightly but significantly elevated even in uninjured Sca-1-/- kidneys. Sca-1 constitutively bound both TGFβ receptors I and II in cultured normal proximal tubular epithelial cells. Its genetic loss or silencing lead to constitutive TGFβ receptor—mediated activation of canonical Smad signaling even in the absence of ligand and to KIM-1 expression in the silenced cells. These studies demonstrate that by normally repressing TGFβ-mediated canonical Smad signaling, Sca-1 plays an important in renal epithelial cell homeostasis and in recovery of renal function following ischemic acute kidney injury.
Collapse
Affiliation(s)
- Troy D. Camarata
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Grant C. Weaver
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Alexandr Vasilyev
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - M. Amin Arnaout
- Leukocyte Biology & Inflammation Program, Renal Division and Department of Medicine Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- Center For Regenerative Medicine, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
24
|
Madera-Sandoval RL, Reyes-Maldonado E, Dzul-Caamal R, Gallegos-Rangel E, Domínguez-López ML, García-Latorre E, Vega-López A. Fat-associated lymphoid cluster in Cyprinus carpio: Characterisation and its relation with peritoneal haemangiosarcoma. FISH & SHELLFISH IMMUNOLOGY 2015; 44:633-641. [PMID: 25804491 DOI: 10.1016/j.fsi.2015.03.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/21/2015] [Accepted: 03/14/2015] [Indexed: 06/04/2023]
Abstract
FALC cells are natural helper cells producing Th2-type cytokines, which express c-kit, Sca-1, IL7R and CD45 in mouse and human. These cells are involved in allergic responses and contribute to the inflammatory reactions of adipose tissue; however, a lack of information prevails about the presence of these cells in other species. The aim of the study was to identify and characterise FALC cells in the common carp (Cyprinus carpio) using immunohistochemistry and molecular biology techniques as well as to explore their relationships with their microenvironment. Histological description of the FALC was performed using H&E and polyclonal antibodies were used against cell-surface markers such as c-kit, Sca-1 and CD45. Furthermore, gene expression of c-kit, Sca-1 and IL7R was assessed. C. carpio FALC cells express the same surface markers reported in FALC of the mouse at both the pre- and post-transcriptional level. By exposure to the soluble fraction of helminths, FALC cells produce abundant Th2 cytokines (IL-5, IL-6 and IL-13) but do not synthesise IL-1α. Additionally, FALC cells probably participate in vascular remodelling of the intestine vessels, inducing tumours because a malignant haemangiosarcoma in the peritoneal cavity was found. In this tumour, abundant FALC with their characteristic cell-surface markers were detected. The findings of this study suggest the involvement of some proto-oncogenes such as c-kit and Sca-1, and the deregulation of Src kinases modulated by CD45 present in C. carpio FALC with the ontogeny of peritoneal haemangiosarcoma in this fish species.
Collapse
Affiliation(s)
- Ruth L Madera-Sandoval
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico
| | - Elba Reyes-Maldonado
- Laboratorio de Citología, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala s/n, Casco de Santo Tomás, México, D.F. CP 11340, Mexico
| | - Ricardo Dzul-Caamal
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico
| | - Esperanza Gallegos-Rangel
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico
| | - María Lilia Domínguez-López
- Laboratorio de Inmunoquímica I, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala s/n, Casco de Santo Tomás, México, D.F. CP 11340, Mexico
| | - Ethel García-Latorre
- Laboratorio de Inmunoquímica I, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Carpio y Plan de Ayala s/n, Casco de Santo Tomás, México, D.F. CP 11340, Mexico
| | - Armando Vega-López
- Laboratorio de Toxicología Ambiental, Departamento de Ingeniería en Sistemas Ambientales, Av. Wilfrido Massieu s/n, Unidad Profesional Zacatenco, México, D.F. CP 07738, Mexico.
| |
Collapse
|
25
|
Song YJ, Choi JH, Lee H. Setdb1 is required for myogenic differentiation of C2C12 myoblast cells via maintenance of MyoD expression. Mol Cells 2015; 38:362-72. [PMID: 25715926 PMCID: PMC4400312 DOI: 10.14348/molcells.2015.2291] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 12/11/2022] Open
Abstract
Setdb1, an H3-K9 specific histone methyltransferase, is associated with transcriptional silencing of euchromatic genes through chromatin modification. Functions of Setdb1 during development have been extensively studied in embryonic and mesenchymal stem cells as well as neurogenic progenitor cells. But the role of Sedtdb1 in myogenic differentiation remains unknown. In this study, we report that Setdb1 is required for myogenic potential of C2C12 myoblast cells through maintaining the expressions of MyoD and muscle-specific genes. We find that reduced Setdb1 expression in C2C12 myoblast cells severely delayed differentiation of C2C12 myoblast cells, whereas exogenous Setdb1 expression had little effect on. Gene expression profiling analysis using oligonucleotide micro-array and RNA-Seq technologies demonstrated that depletion of Setdb1 results in downregulation of MyoD as well as the components of muscle fiber in proliferating C2C12 cells. In addition, exogenous expression of MyoD reversed transcriptional repression of MyoD promoter-driven lucif-erase reporter by Setdb1 shRNA and rescued myogenic differentiation of C2C12 myoblast cells depleted of endogenous Setdb1. Taken together, these results provide new insights into how levels of key myogenic regulators are maintained prior to induction of differentiation.
Collapse
Affiliation(s)
- Young Joon Song
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon 402-751,
Korea
| | - Jang Hyun Choi
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon 402-751,
Korea
| | - Hansol Lee
- Department of Biological Sciences, College of Natural Science, Inha University, Incheon 402-751,
Korea
| |
Collapse
|
26
|
Bogdan P, Deasy BM, Gharaibeh B, Roehrs T, Marculescu R. Heterogeneous structure of stem cells dynamics: statistical models and quantitative predictions. Sci Rep 2014; 4:4826. [PMID: 24769917 PMCID: PMC4001100 DOI: 10.1038/srep04826] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 04/08/2014] [Indexed: 01/08/2023] Open
Abstract
Understanding stem cell (SC) population dynamics is essential for developing models that can be used in basic science and medicine, to aid in predicting cells fate. These models can be used as tools e.g. in studying patho-physiological events at the cellular and tissue level, predicting (mal)functions along the developmental course, and personalized regenerative medicine. Using time-lapsed imaging and statistical tools, we show that the dynamics of SC populations involve a heterogeneous structure consisting of multiple sub-population behaviors. Using non-Gaussian statistical approaches, we identify the co-existence of fast and slow dividing subpopulations, and quiescent cells, in stem cells from three species. The mathematical analysis also shows that, instead of developing independently, SCs exhibit a time-dependent fractal behavior as they interact with each other through molecular and tactile signals. These findings suggest that more sophisticated models of SC dynamics should view SC populations as a collective and avoid the simplifying homogeneity assumption by accounting for the presence of more than one dividing sub-population, and their multi-fractal characteristics.
Collapse
Affiliation(s)
- Paul Bogdan
- 1] Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089-2560, USA [2]
| | - Bridget M Deasy
- 1] CellStock, Pittsburgh, PA 15237, USA [2] McGowan Institute of Regenerative Medicine of UPMC and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA [3]
| | - Burhan Gharaibeh
- 1] Institute for Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, PA15213, USA [2] Stem Cell Research Center (SCRC), University of Pittsburgh, Pittsburgh, PA 15219, USA [3]
| | - Timo Roehrs
- McGowan Institute of Regenerative Medicine of UPMC and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Radu Marculescu
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
He WA, Berardi E, Cardillo VM, Acharyya S, Aulino P, Thomas-Ahner J, Wang J, Bloomston M, Muscarella P, Nau P, Shah N, Butchbach MER, Ladner K, Adamo S, Rudnicki MA, Keller C, Coletti D, Montanaro F, Guttridge DC. NF-κB-mediated Pax7 dysregulation in the muscle microenvironment promotes cancer cachexia. J Clin Invest 2014; 123:4821-35. [PMID: 24084740 DOI: 10.1172/jci68523] [Citation(s) in RCA: 274] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 08/06/2013] [Indexed: 01/09/2023] Open
Abstract
Cachexia is a debilitating condition characterized by extreme skeletal muscle wasting that contributes significantly to morbidity and mortality. Efforts to elucidate the underlying mechanisms of muscle loss have predominantly focused on events intrinsic to the myofiber. In contrast, less regard has been given to potential contributory factors outside the fiber within the muscle microenvironment. In tumor-bearing mice and patients with pancreatic cancer, we found that cachexia was associated with a type of muscle damage resulting in activation of both satellite and nonsatellite muscle progenitor cells. These muscle progenitors committed to a myogenic program, but were inhibited from completing differentiation by an event linked with persistent expression of the self-renewing factor Pax7. Overexpression of Pax7 was sufficient to induce atrophy in normal muscle, while under tumor conditions, the reduction of Pax7 or exogenous addition of its downstream target, MyoD, reversed wasting by restoring cell differentiation and fusion with injured fibers. Furthermore, Pax7 was induced by serum factors from cachectic mice and patients, in an NF-κB-dependent manner, both in vitro and in vivo. Together, these results suggest that Pax7 responds to NF-κB by impairing the regenerative capacity of myogenic cells in the muscle microenvironment to drive muscle wasting in cancer.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Animals
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Case-Control Studies
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Mice, Inbred mdx
- Mice, Nude
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Middle Aged
- Muscle Development
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- NF-kappa B/metabolism
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Pancreatic Neoplasms/complications
- Pancreatic Neoplasms/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Tumor Microenvironment
- Young Adult
Collapse
|
28
|
Hsu YC, Mildenstein K, Hunter K, Tkachenko O, Mullen CA. Acute lymphoid leukemia cells with greater stem cell antigen-1 (Ly6a/Sca-1) expression exhibit higher levels of metalloproteinase activity and are more aggressive in vivo. PLoS One 2014; 9:e88966. [PMID: 24586463 PMCID: PMC3930640 DOI: 10.1371/journal.pone.0088966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/16/2014] [Indexed: 12/13/2022] Open
Abstract
Stem cell antigen-1 (Ly6a/Sca-1) is a gene that is expressed in activated lymphocytes, hematopoietic stem cells and stem cells of a variety of tissues in mice. Despite decades of study its functions remain poorly defined. These studies explored the impact of expression of this stem cell associated gene in acute lymphoid leukemia. Higher levels of Ly6a/Sca-1 expression led to more aggressive leukemia growth in vivo and earlier death of hosts. Leukemias expressing higher levels of Ly6a/Sca-1 exhibited higher levels of matrix metalloproteinases. The results suggest the hypothesis that the more aggressive behavior of Ly6a/Sca-1 expressing leukemias is due at least in part to greater capacity to degrade microenvironmental stroma and invade tissues.
Collapse
Affiliation(s)
- Yu-Chiao Hsu
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Kurt Mildenstein
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Kordell Hunter
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Olena Tkachenko
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
| | - Craig A. Mullen
- Department of Pediatrics, University of Rochester, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
29
|
Azrak SS, Ginel-Picardo A, Drosten M, Barbacid M, Santos E. Reversible, interrelated mRNA and miRNA expression patterns in the transcriptome of Rasless fibroblasts: functional and mechanistic implications. BMC Genomics 2013; 14:731. [PMID: 24156637 PMCID: PMC4007593 DOI: 10.1186/1471-2164-14-731] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/04/2013] [Indexed: 12/13/2022] Open
Abstract
Background 4-Hydroxy-tamoxifen (4OHT) triggers Cre-mediated K-Ras removal in [H-Ras-/-;N-Ras-/-;K-Raslox/lox;RERTert/ert] fibroblasts, generating growth-arrested “Rasless” MEFs which are able to recover their proliferative ability after ectopic expression of Ras oncoproteins or constitutively active BRAF or MEK1. Results Comparison of the transcriptional profiles of Rasless fibroblasts with those of MEFs lacking only H-Ras and N-Ras identified a series of differentially expressed mRNAs and microRNAs specifically linked to the disappearance of K-Ras from these cells. The rescue of cell cycle progression in Rasless cells by activated BRAF or MEK1 resulted in the reversal of most such transcriptional mRNA and microRNA alterations. Functional analysis of the differentially expressed mRNAs uncovered a significant enrichment in the components of pathways regulating cell division, DNA/RNA processing and response to DNA damage. Consistent with G1/S blockade, Rasless cells displayed repression of a series of cell cycle-related genes, including Cyclins, Cyclin-dependent kinases, Myc and E2F transcription targets, and upregulation of Cyclin-dependent kinase inhibitors. The profile of differentially expressed microRNAs included a specific set of oncomiR families and clusters (repressed miR-17 ~ 92, miR-106a ~ 363, miR-106b ~ 25, miR-212 ~ 132, miR-183 ~ 182, and upregulated miR-335) known for their ability to target a specific set of cellular regulators and checkpoint sensors (including Rb, E2F and Cdkns) able to modulate the interplay between the pro- and anti-proliferative or stress-response pathways that are reversibly altered in Rasless cells. Conclusions Our data suggest that the reversible proliferation phenotype of Rasless cells is the pleiotropic result of interplay among distinct pro- and anti-proliferative, and stress-response pathways modulated by a regulatory circuitry constituted by a specific set of differentially expressed mRNAs and microRNAs and preferentially targeting two cross-talking signalling axes: Myc-Rb-E2F-dependent and Cdkns-p53-dependent pathways.
Collapse
Affiliation(s)
| | | | | | | | - Eugenio Santos
- Centro de Investigacion del Cancer, IBMCC (CSIC-USAL), University of Salamanca, Campus Unamuno, 37007, Salamanca, Spain.
| |
Collapse
|
30
|
Ancestral Myf5 gene activity in periocular connective tissue identifies a subset of fibro/adipogenic progenitors but does not connote a myogenic origin. Dev Biol 2013; 385:366-79. [PMID: 23969310 DOI: 10.1016/j.ydbio.2013.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/21/2013] [Accepted: 08/13/2013] [Indexed: 11/23/2022]
Abstract
Extraocular muscles (EOM) represent a unique muscle group that controls eye movements and originates from head mesoderm, while the more typically studied body and limb muscles are somite-derived. Aiming to investigate myogenic progenitors (satellite cells) in EOM versus limb and diaphragm of adult mice, we have been using flow cytometry in combination with myogenic-specific Cre-loxP lineage marking for cell isolation. While analyzing cells from the EOM of mice that harbor Myf5(Cre)-driven GFP expression, we identified in addition to the expected GFP(+) myogenic cells (presumably satellite cells), a second dominant GFP(+) population distinguished as being Sca1(+), non-myogenic, and exhibiting a fibro/adipogenic potential. This unexpected population was not only unique to EOM compared to the other muscles but also specific to the Myf5(Cre)-driven reporter when compared to the MyoD(Cre) driver. Histological studies of periocular tissue preparations demonstrated the presence of Myf5(Cre)-driven GFP(+) cells in connective tissue locations adjacent to the muscle masses, including cells in the vasculature wall. These vasculature-associated GFP(+) cells were further identified as mural cells based on the presence of the specific XLacZ4 transgene. Unlike the EOM satellite cells that originate from a Pax3-negative lineage, these non-myogenic Myf5(Cre)-driven GFP(+) cells appear to be related to cells of a Pax3-expressing origin, presumably derived from the neural crest. In all, our lineage tracing based on multiple reporter lines has demonstrated that regardless of common ancestral expression of Myf5, there is a clear distinction between periocular myogenic and non-myogenic cell lineages according to their mutually exclusive antecedence of MyoD and Pax3 gene activity.
Collapse
|
31
|
Bernstein HS, Samad T, Cholsiripunlert S, Khalifian S, Gong W, Ritner C, Aurigui J, Ling V, Wilschut KJ, Bennett S, Hoffman J, Oishi P. Stem cell antigen-1 in skeletal muscle function. PLOS CURRENTS 2013; 5. [PMID: 24042315 PMCID: PMC3770837 DOI: 10.1371/currents.md.411a8332d61e22725e6937b97e6d0ef8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Stem cell antigen-1 (Sca-1) is a member of the Ly-6 multigene family encoding highly homologous, glycosyl-phosphatidylinositol-anchored membrane proteins. Sca-1 is expressed on muscle-derived stem cells and myogenic precursors recruited to sites of muscle injury. We previously reported that inhibition of Sca-1 expression stimulated myoblast proliferation in vitro and regulated the tempo of muscle repair in vivo. Despite its function in myoblast expansion during muscle repair, a role for Sca-1 in normal, post-natal muscle has not been thoroughly investigated. We systematically compared Sca-1-/- (KO) and Sca-1+/+ (WT) mice and hindlimb muscles to elucidate the tissue, contractile, and functional effects of Sca-1 in young and aging animals. Comparison of muscle volume, fibrosis, myofiber cross-sectional area, and Pax7+ myoblast number showed little differences between ages or genotypes. Exercise protocols, however, demonstrated decreased stamina in KO versus WT mice, with young KO mice achieving results similar to aging WT animals. In addition, KO mice did not improve with practice, while WT animals demonstrated conditioning over time. Surprisingly, myomechanical analysis of isolated muscles showed that KO young muscle generated more force and experienced less fatigue. However, KO muscle also demonstrated incomplete relaxation with fatigue. These findings suggest that Sca-1 is necessary for muscle conditioning with exercise, and that deficient conditioning in Sca-1 KO animals becomes more pronounced with age.
Collapse
|
32
|
Sakai H, Sato T, Sakurai H, Yamamoto T, Hanaoka K, Montarras D, Sehara-Fujisawa A. Fetal skeletal muscle progenitors have regenerative capacity after intramuscular engraftment in dystrophin deficient mice. PLoS One 2013; 8:e63016. [PMID: 23671652 PMCID: PMC3650009 DOI: 10.1371/journal.pone.0063016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/27/2013] [Indexed: 12/13/2022] Open
Abstract
Muscle satellite cells (SCs) are stem cells that reside in skeletal muscles and contribute to regeneration upon muscle injury. SCs arise from skeletal muscle progenitors expressing transcription factors Pax3 and/or Pax7 during embryogenesis in mice. However, it is unclear whether these fetal progenitors possess regenerative ability when transplanted in adult muscle. Here we address this question by investigating whether fetal skeletal muscle progenitors (FMPs) isolated from Pax3GFP/+ embryos have the capacity to regenerate muscle after engraftment into Dystrophin-deficient mice, a model of Duchenne muscular dystrophy. The capacity of FMPs to engraft and enter the myogenic program in regenerating muscle was compared with that of SCs derived from adult Pax3GFP/+ mice. Transplanted FMPs contributed to the reconstitution of damaged myofibers in Dystrophin-deficient mice. However, despite FMPs and SCs having similar myogenic ability in culture, the regenerative ability of FMPs was less than that of SCs in vivo. FMPs that had activated MyoD engrafted more efficiently to regenerate myofibers than MyoD-negative FMPs. Transcriptome and surface marker analyses of these cells suggest the importance of myogenic priming for the efficient myogenic engraftment. Our findings suggest the regenerative capability of FMPs in the context of muscle repair and cell therapy for degenerative muscle disease.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Dystrophin/deficiency
- Dystrophin/genetics
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Immunohistochemistry
- Injections, Intramuscular
- Mice
- Mice, Knockout
- Mice, Transgenic
- Muscle, Skeletal/cytology
- Muscle, Skeletal/embryology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/physiopathology
- Muscular Dystrophy, Animal/surgery
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/surgery
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/transplantation
- Myofibrils/genetics
- Myofibrils/physiology
- Myogenin/genetics
- Myogenin/metabolism
- PAX3 Transcription Factor
- Paired Box Transcription Factors/genetics
- Paired Box Transcription Factors/metabolism
- Regeneration/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Satellite Cells, Skeletal Muscle/transplantation
- Stem Cell Transplantation/methods
- Transcriptome
Collapse
Affiliation(s)
- Hiroshi Sakai
- Department of Growth Regulation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Takahiko Sato
- Department of Growth Regulation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- * E-mail: (TS); (AS-F)
| | - Hidetoshi Sakurai
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Reprogramming Science, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Kazunori Hanaoka
- Laboratory of Molecular Embryology, Department of Bioscience, Kitasato University School of Science, Kanagawa, Japan
| | - Didier Montarras
- Molecular Genetics of Development, Institut Pasteur, Paris, France
| | - Atsuko Sehara-Fujisawa
- Department of Growth Regulation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- * E-mail: (TS); (AS-F)
| |
Collapse
|
33
|
Mozzetta C, Consalvi S, Saccone V, Tierney M, Diamantini A, Mitchell KJ, Marazzi G, Borsellino G, Battistini L, Sassoon D, Sacco A, Puri PL. Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice. EMBO Mol Med 2013; 5:626-39. [PMID: 23505062 PMCID: PMC3628105 DOI: 10.1002/emmm.201202096] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 02/02/2013] [Accepted: 02/04/2013] [Indexed: 12/26/2022] Open
Abstract
HDAC inhibitors (HDACi) exert beneficial effects in mdx mice, by promoting endogenous regeneration; however, the cellular determinants of HDACi activity on dystrophic muscles have not been determined. We show that fibroadipogenic progenitors (FAP) influence the regeneration potential of satellite cells during disease progression in mdx mice and mediate HDACi ability to selectively promote regeneration at early stages of disease. FAPs from young mdx mice promote, while FAPs from old mdx mice repress, satellite cell-mediated formation of myotubes. In young mdx mice HDACi inhibited FAP adipogenic potential, while enhancing their ability to promote differentiation of adjacent satellite cells, through upregulation of the soluble factor follistatin. By contrast, FAPs from old mdx mice were resistant to HDACi-mediated inhibition of adipogenesis and constitutively repressed satellite cell-mediated formation of myotubes. We show that transplantation of FAPs from regenerating young muscles restored HDACi ability to increase myofibre size in old mdx mice. These results reveal that FAPs are key cellular determinants of disease progression in mdx mice and mediate a previously unappreciated stage-specific beneficial effect of HDACi in dystrophic muscles.
Collapse
|
34
|
Chapman MR, Balakrishnan KR, Li J, Conboy MJ, Huang H, Mohanty SK, Jabart E, Hack J, Conboy IM, Sohn LL. Sorting single satellite cells from individual myofibers reveals heterogeneity in cell-surface markers and myogenic capacity. Integr Biol (Camb) 2013; 5:692-702. [PMID: 23407661 DOI: 10.1039/c3ib20290a] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traditional cell-screening techniques such as FACS and MACS are better suited for large numbers of cells isolated from bulk tissue and cannot easily screen stem or progenitor cells from minute populations found in their physiological niches. Furthermore, these techniques rely upon irreversible antibody binding, potentially altering cell properties, including gene expression and regenerative capacity. To address these challenges, we have developed a novel, label-free stem-cell analysis and sorting platform capable of quantifying cell-surface marker expression of single functional organ stem cells directly isolated from their micro-anatomical niche. Using our unique platform, we have discovered a remarkable heterogeneity in both the regenerative capacity and expression of CXCR4, β1-integrin, Sca-1, M-cadherin, Syndecan-4, and Notch-1 in freshly isolated muscle stem (satellite) cells residing on different, single myofibers and have identified a small population of Sca-1(+)/Myf5(+) myogenic satellite cells. Our results demonstrate the utility of our single-cell platform for uncovering and functionally characterizing stem-cell heterogeneity in the organ microniche.
Collapse
Affiliation(s)
- Matthew R Chapman
- Biophysics Graduate Group, University of California, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Role of akirin in skeletal myogenesis. Int J Mol Sci 2013; 14:3817-23. [PMID: 23396110 PMCID: PMC3588072 DOI: 10.3390/ijms14023817] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 01/23/2013] [Accepted: 01/31/2013] [Indexed: 11/17/2022] Open
Abstract
Akirin is a recently discovered nuclear factor that plays an important role in innate immune responses. Beyond its role in innate immune responses, Akirin has recently been shown to play an important role in skeletal myogenesis. In this article, we will briefly review the structure and tissue distribution of Akirin and discuss recent advances in our understanding of its role and signal pathway in skeletal myogenesis.
Collapse
|
36
|
Penton CM, Thomas-Ahner JM, Johnson EK, McAllister C, Montanaro F. Muscle side population cells from dystrophic or injured muscle adopt a fibro-adipogenic fate. PLoS One 2013; 8:e54553. [PMID: 23336007 PMCID: PMC3545954 DOI: 10.1371/journal.pone.0054553] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 12/14/2012] [Indexed: 11/17/2022] Open
Abstract
Muscle side population (SP) cells are rare multipotent stem cells that can participate in myogenesis and muscle regeneration upon transplantation. While they have been primarily studied for the development of cell-based therapies for Duchenne muscular dystrophy, little is known regarding their non-muscle lineage choices or whether the dystrophic muscle environment affects their ability to repair muscle. Unfortunately, the study of muscle SP cells has been challenged by their low abundance and the absence of specific SP cell markers. To address these issues, we developed culture conditions for the propagation and spontaneous multi-lineage differentiation of muscle SP cells. Using this approach, we show that SP cells from wild type muscle robustly differentiate into satellite cells and form myotubes without requiring co-culture with myogenic cells. Furthermore, this myogenic activity is associated with SP cells negative for immune (CD45) and vascular (CD31) markers but positive for Pax7, Sca1, and the mesenchymal progenitor marker PDGFRα. Additionally, our studies revealed that SP cells isolated from dystrophic or cardiotoxin-injured muscle fail to undergo myogenesis. Instead, these SP cells rapidly expand giving rise to fibroblast and adipocyte progenitors (FAPs) and to their differentiated progeny, fibroblasts and adipocytes. Our findings indicate that muscle damage affects the lineage choices of muscle SP cells, promoting their differentiation along fibro-adipogenic lineages while inhibiting myogenesis. These results have implications for a possible role of muscle SP cells in fibrosis and fat deposition in muscular dystrophy. In addition, our studies provide a useful in vitro system to analyze SP cell biology in both normal and pathological conditions.
Collapse
Affiliation(s)
- Christopher M Penton
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
37
|
Bailey B, Fransioli J, Gude NA, Alvarez R, Zhang X, Zhan X, Gustafsson ÅB, Sussman MA. Sca-1 knockout impairs myocardial and cardiac progenitor cell function. Circ Res 2012; 111:750-60. [PMID: 22800687 DOI: 10.1161/circresaha.112.274662] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
RATIONALE Cardiac progenitor cells are important for maintenance of myocardial structure and function, but molecular mechanisms governing these progenitor cells remain obscure and require elucidation to enhance regenerative therapeutic approaches. OBJECTIVE To understand consequences of stem cell antigen-1 (Sca-1) deletion on functional properties of c-kit+ cardiac progenitor cells and myocardial performance using a Sca-1 knock-out/green fluorescent protein knock-in reporter mouse (ScaKI). METHODS AND RESULTS Genetic deletion of Sca-1 results in early-onset cardiac contractile deficiency as determined by echocardiography and hemodynamics as well as age-associated hypertrophy. Resident cardiac progenitor cells in ScaKI mice do not respond to pathological damage in vivo, consistent with observations of impaired growth and survival of ScaKI cardiac progenitor cells in vitro. The molecular basis of the defect in ScaKI cardiac progenitor cells is associated with increased canonical Wnt signaling pathway activation consistent with molecular characteristics of lineage commitment. CONCLUSIONS Genetic deletion of Sca-1 causes primary cardiac defects in myocardial contractility and repair consistent with impairment of resident cardiac progenitor cell proliferative capacity associated with altered canonical Wnt signaling.
Collapse
Affiliation(s)
- Brandi Bailey
- SDSU Heart Institute, and Department of Biology, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Penvose A, Westerman KA. Sca-1 is involved in the adhesion of myosphere cells to αVβ3 integrin. Biol Open 2012; 1:839-47. [PMID: 23213478 PMCID: PMC3507234 DOI: 10.1242/bio.20121222] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 06/07/2012] [Indexed: 12/18/2022] Open
Abstract
A myosphere cell is a unique type of muscle stem cell that is able to maintain its pre-myogenic state in culture over time. These cells are propagated in culture as free-floating, non-adherent spheres. We believe that the 3-dimensional adhesive cell-cell interactions involved in maintaining the sphere-like myosphere structures are also involved in maintaining their longevity in culture. We found that Sca-1, which is highly expressed by myosphere cells, plays a role in the growth and the formation of the myospheres. In comparing adhesion molecules expressed by 3-dimensionally grown myosphere cells to those expressed by 2-dimensionally grown primary myoblasts, we found that there was a distinct difference in the expression of β3 integrin. Upon further investigation we discovered that there is an adhesive interaction between Sca-1(+) cells and αVβ3 integrin. Here we show that Sca-1(+) cells (myosphere cells and NIH3T3 cells) adhere to αVβ3 integrin and that Sca-1(-) cells (primary myoblasts) do not adhere. The interaction between Sca-1 and αVβ3 integrin was confirmed using antibody blocking, shRNA knockdown of Sca-1 in Sca-1(+) cells, and by expressing Sca-1 cDNA in Sca-1(-) cells, which demonstrated that the level of adhesion of these cells to αVβ3 integrin was dependent on the presence of Sca-1. Additionally, we found that the co-expression of Sca-1 and β3 resulted in significantly greater adhesion of Sca-1(+) cells to αVβ3 integrin. In conclusion, our data indicate that Sca-1 is involved in maintaining the 3-dimensional myosphere cell-cell contacts and that Sca-1 is involved in the binding of cells to αVβ3 integrin.
Collapse
Affiliation(s)
- Ashley Penvose
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women's Hospital , 75 Francis Street, Boston, MA 02115 , USA
| | | |
Collapse
|
39
|
Rocheteau P, Gayraud-Morel B, Siegl-Cachedenier I, Blasco MA, Tajbakhsh S. A subpopulation of adult skeletal muscle stem cells retains all template DNA strands after cell division. Cell 2012; 148:112-25. [PMID: 22265406 DOI: 10.1016/j.cell.2011.11.049] [Citation(s) in RCA: 381] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 10/20/2011] [Accepted: 11/03/2011] [Indexed: 12/31/2022]
Abstract
Satellite cells are adult skeletal muscle stem cells that are quiescent and constitute a poorly defined heterogeneous population. Using transgenic Tg:Pax7-nGFP mice, we show that Pax7-nGFP(Hi) cells are less primed for commitment and have a lower metabolic status and delayed first mitosis compared to Pax7-nGFP(Lo) cells. Pax7-nGFP(Hi) can give rise to Pax7-nGFP(Lo) cells after serial transplantations. Proliferating Pax7-nGFP(Hi) cells exhibit lower metabolic activity, and the majority performs asymmetric DNA segregation during cell division, wherein daughter cells retaining template DNA strands express stem cell markers. Using chromosome orientation-fluorescence in situ hybridization, we demonstrate that all chromatids segregate asymmetrically, whereas Pax7-nGFP(Lo) cells perform random DNA segregation. Therefore, quiescent Pax7-nGFP(Hi) cells represent a reversible dormant stem cell state, and during muscle regeneration, Pax7-nGFP(Hi) cells generate distinct daughter cell fates by asymmetrically segregating template DNA strands to the stem cell. These findings provide major insights into the biology of stem cells that segregate DNA asymmetrically.
Collapse
Affiliation(s)
- Pierre Rocheteau
- Institut Pasteur, Stem Cells and Development, Department of Developmental Biology, CNRS URA 2578, 25 rue du Dr. Roux, Paris 75015, France
| | | | | | | | | |
Collapse
|
40
|
Valero MC, Huntsman HD, Liu J, Zou K, Boppart MD. Eccentric exercise facilitates mesenchymal stem cell appearance in skeletal muscle. PLoS One 2012; 7:e29760. [PMID: 22253772 PMCID: PMC3256189 DOI: 10.1371/journal.pone.0029760] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 12/03/2011] [Indexed: 01/13/2023] Open
Abstract
Eccentric, or lengthening, contractions result in injury and subsequently stimulate the activation and proliferation of satellite stem cells which are important for skeletal muscle regeneration. The discovery of alternative myogenic progenitors in skeletal muscle raises the question as to whether stem cells other than satellite cells accumulate in muscle in response to exercise and contribute to post-exercise repair and/or growth. In this study, stem cell antigen-1 (Sca-1) positive, non-hematopoetic (CD45-) cells were evaluated in wild type (WT) and α7 integrin transgenic (α7Tg) mouse muscle, which is resistant to injury yet liable to strain, 24 hr following a single bout of eccentric exercise. Sca-1+CD45− stem cells were increased 2-fold in WT muscle post-exercise. The α7 integrin regulated the presence of Sca-1+ cells, with expansion occurring in α7Tg muscle and minimal cells present in muscle lacking the α7 integrin. Sca-1+CD45− cells isolated from α7Tg muscle following exercise were characterized as mesenchymal-like stem cells (mMSCs), predominantly pericytes. In vitro multiaxial strain upregulated mMSC stem cells markers in the presence of laminin, but not gelatin, identifying a potential mechanistic basis for the accumulation of these cells in muscle following exercise. Transplantation of DiI-labeled mMSCs into WT muscle increased Pax7+ cells and facilitated formation of eMHC+DiI− fibers. This study provides the first demonstration that mMSCs rapidly appear in skeletal muscle in an α7 integrin dependent manner post-exercise, revealing an early event that may be necessary for effective repair and/or growth following exercise. The results from this study also support a role for the α7 integrin and/or mMSCs in molecular- and cellular-based therapeutic strategies that can effectively combat disuse muscle atrophy.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Ataxin-1
- Ataxins
- Biomarkers/metabolism
- Cell Proliferation/drug effects
- Cell Separation
- Connective Tissue Cells/cytology
- Female
- Gelatin/pharmacology
- Integrin alpha Chains/metabolism
- Laminin/metabolism
- Leukocyte Common Antigens/metabolism
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/drug effects
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Transgenic
- Multipotent Stem Cells/cytology
- Multipotent Stem Cells/drug effects
- Muscle Development/drug effects
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Nerve Tissue Proteins/metabolism
- Nuclear Proteins/metabolism
- PAX7 Transcription Factor/metabolism
- Pericytes/cytology
- Pericytes/drug effects
- Physical Conditioning, Animal
- Stem Cell Transplantation
- Stress, Mechanical
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- M. Carmen Valero
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Heather D. Huntsman
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Jianming Liu
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Kai Zou
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
| | - Marni D. Boppart
- Department of Kinesiology and Community Health, and Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
41
|
Muscle-derived stem/progenitor cell dysfunction limits healthspan and lifespan in a murine progeria model. Nat Commun 2012; 3:608. [PMID: 22215083 PMCID: PMC3272577 DOI: 10.1038/ncomms1611] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 11/24/2011] [Indexed: 01/04/2023] Open
Abstract
With ageing, there is a loss of adult stem cell function. However, there is no direct evidence that this has a causal role in ageing-related decline. We tested this using muscle-derived stem/progenitor cells (MDSPCs) in a murine progeria model. Here we show that MDSPCs from old and progeroid mice are defective in proliferation and multilineage differentiation. Intraperitoneal administration of MDSPCs, isolated from young wild-type mice, to progeroid mice confer significant lifespan and healthspan extension. The transplanted MDSPCs improve degenerative changes and vascularization in tissues where donor cells are not detected, suggesting that their therapeutic effect may be mediated by secreted factor(s). Indeed, young wild-type-MDSPCs rescue proliferation and differentiation defects of aged MDSPCs when co-cultured. These results establish that adult stem/progenitor cell dysfunction contributes to ageing-related degeneration and suggests a therapeutic potential of post-natal stem cells to extend health. The function of adult stem cells is diminished with age but the role this dysfunction plays in the aging process is unknown. Here, the injection of muscle-derived stem/progenitor cells from young mice rescues symptoms in progeroid mice and is shown to regenerate tissues independent of engraftment.
Collapse
|
42
|
Yuan H, Upadhyay G, Yin Y, Kopelovich L, Glazer RI. Stem cell antigen-1 deficiency enhances the chemopreventive effect of peroxisome proliferator-activated receptorγ activation. Cancer Prev Res (Phila) 2012; 5:51-60. [PMID: 21955520 PMCID: PMC3252486 DOI: 10.1158/1940-6207.capr-11-0256] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Stem cell antigen-1 (Sca-1, Ly6A) is a glycerophosphatidylinositol (GPI)-anchored protein that was identified as a murine marker of bone marrow stem cells. Although Sca-1 is widely used to enrich for stem and progenitor cells in various tissues, little is known about its function and associated signaling pathways in normal and malignant cells. Here, we report that the absence of Sca-1 in the mammary gland resulted in higher levels of PPARγ and PTEN, and a reduction of pSer84PPARγ, pERK1/2, and PPARδ. This phenotype correlated with markedly increased sensitivity of Sca-1 null mice to PPARγ agonist GW7845 and insensitivity to PPARδ agonist GW501516. Reduction of Sca-1 expression in mammary tumor cells by RNA interference resulted in a phenotype similar to the Sca-1 deficient mammary gland, as evidenced by increased PPARγ expression and transcriptional activity, resulting in part from a lesser susceptibility to proteasomal degradation. These data implicate Sca-1 as a negative regulator of the tumor suppressor effects of PPARγ.
Collapse
Affiliation(s)
- Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Geeta Upadhyay
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Yuzhi Yin
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Levy Kopelovich
- Chemoprevention Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Robert I. Glazer
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
43
|
Doyle MJ, Zhou S, Tanaka KK, Pisconti A, Farina NH, Sorrentino BP, Olwin BB. Abcg2 labels multiple cell types in skeletal muscle and participates in muscle regeneration. ACTA ACUST UNITED AC 2011; 195:147-63. [PMID: 21949413 PMCID: PMC3187700 DOI: 10.1083/jcb.201103159] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abcg2-expressing cells proliferate after muscle injury and are required for effective regeneration of multiple muscle cell lineages. Skeletal muscle contains progenitor cells (satellite cells) that maintain and repair muscle. It also contains muscle side population (SP) cells, which express Abcg2 and may participate in muscle regeneration or may represent a source of satellite cell replenishment. In Abcg2-null mice, the SP fraction is lost in skeletal muscle, although the significance of this loss was previously unknown. We show that cells expressing Abcg2 increased upon injury and that muscle regeneration was impaired in Abcg2-null mice, resulting in fewer centrally nucleated myofibers, reduced myofiber size, and fewer satellite cells. Additionally, using genetic lineage tracing, we demonstrate that the progeny of Abcg2-expressing cells contributed to multiple cell types within the muscle interstitium, primarily endothelial cells. After injury, Abcg2 progeny made a minor contribution to regenerated myofibers. Furthermore, Abcg2-labeled cells increased significantly upon injury and appeared to traffic to muscle from peripheral blood. Together, these data suggest an important role for Abcg2 in positively regulating skeletal muscle regeneration.
Collapse
Affiliation(s)
- Michelle J Doyle
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Stem cell antigen-1 enhances tumorigenicity by disruption of growth differentiation factor-10 (GDF10)-dependent TGF-beta signaling. Proc Natl Acad Sci U S A 2011; 108:7820-5. [PMID: 21518866 DOI: 10.1073/pnas.1103441108] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Stem cell antigen (Sca)-1/Ly6A, a glycerophosphatidylinositol-linked surface protein, was found to be associated with murine stem cell- and progenitor cell-enriched populations, and also has been linked to the capacity of tumor-initiating cells. Despite these interesting associations, this protein's functional role in these processes remains largely unknown. To identify the mechanism underlying the protein's possible role in mammary tumorigenesis, Sca-1 expression was examined in Sca-1(+/EGFP) mice during carcinogenesis. Mammary tumor cells derived from these mice readily engrafted in syngeneic mice, and tumor growth was markedly inhibited on down-regulation of Sca-1 expression. The latter effect was associated with significantly elevated expression of the TGF-β ligand growth differentiation factor-10 (GDF10), which was found to selectively activate TGF-β receptor (TβRI/II)-dependent Smad3 phosphorylation. Overexpression of GDF10 attenuated tumor formation; conversely, silencing of GDF10 expression reversed these effects. Sca-1 attenuated GDF10-dependent TGF-β signaling by disrupting the heterodimerization of TβRI and TβRII receptors. These findings suggest a new functional role for Sca-1 in maintaining tumorigenicity, in part by acting as a potent suppressor of TGF-β signaling.
Collapse
|
45
|
Purwanti N, Tsuji D, Azlina A, Karabasil MR, Javkhlan P, Hasegawa T, Yao C, Akamatsu T, Itoh K, Hosoi K. Induction of Sca-1 in the duct cells of the mouse submandibular gland by obstruction of the main excretory duct. J Oral Pathol Med 2011; 40:651-8. [PMID: 21884259 DOI: 10.1111/j.1600-0714.2011.01011.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The effect of ligation of the main excretory duct (MED) of the mouse submandibular gland (SMG) on the expression of Sca-1, a stem cell antigen, was examined by Western blotting and immunohistochemistry. By Western blotting, the expression of Sca-1 with a molecular weight of 18 kDa was identified in the normal gland. At 1 day post-ligation, the expression level of Sca-1 was strongly increased in the experimental gland and weakly in the contralateral gland, and such expression in both glands decreased at 6 days. By immunohistochemistry, Sca-1 was detected weakly in the apical membrane of excretory duct (ED) cells of the SMG under the normal condition. By duct ligation, Sca-1 became expressed strongly in most cells of the two major duct systems, i.e., the striated duct (SD) and granular convoluted tubules (GCT), but was not detected in the acinar (Ac) cells. By fluorescence-activated cell sorter (FACS) analysis, the number of side population (SP) cells in this gland was found to be increased by ligation. These results imply that Sca-1-positive cells may have a role in the duct cell proliferation in the regeneration step elicited by MED ligation-induced injury.
Collapse
Affiliation(s)
- Nunuk Purwanti
- Department of Molecular Oral Physiology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima-shi, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Long KK, Montano M, Pavlath GK. Sca-1 is negatively regulated by TGF-beta1 in myogenic cells. FASEB J 2010; 25:1156-65. [PMID: 21156809 DOI: 10.1096/fj.10-170308] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sca-1 (stem cell antigen-1) is a member of the Ly-6 family of proteins and regulates cell proliferation, differentiation, and self-renewal in multiple tissues. In skeletal muscle, Sca-1 inhibits both proliferation and differentiation of myogenic cells. Sca-1 expression is dynamically regulated during muscle regeneration, and mice lacking Sca-1 display increased fibrosis following muscle injury. Here, we show that Sca-1 expression is negatively regulated by TGF-β1 and that this inhibition is dependent on Smad3. We demonstrate that levels of TGF-β1 in skeletal muscle rapidly increase on injury and that the majority of this TGFβ1 is produced by infiltrating macrophages. Sca-1 is expressed in multiple cell types, and we demonstrate that TGF-β1 represses Sca-1 expression in T cells and other immune cell populations derived from the spleen, indicating that regulation by TGF-β1 is a general feature of Sca-1 expression in multiple cell types. Elucidation of the mechanisms by which Sca-1 expression is regulated may aid in the understanding of muscle homeostasis, potentially identifying novel therapeutic targets for muscle diseases.
Collapse
Affiliation(s)
- Kimberly K Long
- Department of Infectious Diseases, Boston Medical Center, Boston, Massachusetts, USA
| | | | | |
Collapse
|
47
|
Boldrin L, Muntoni F, Morgan JE. Are human and mouse satellite cells really the same? J Histochem Cytochem 2010; 58:941-55. [PMID: 20644208 PMCID: PMC2958137 DOI: 10.1369/jhc.2010.956201] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 06/24/2010] [Indexed: 12/20/2022] Open
Abstract
Satellite cells are quiescent cells located under the basal lamina of skeletal muscle fibers that contribute to muscle growth, maintenance, repair, and regeneration. Mouse satellite cells have been shown to be muscle stem cells that are able to regenerate muscle fibers and self-renew. As human skeletal muscle is also able to regenerate following injury, we assume that the human satellite cell is, like its murine equivalent, a muscle stem cell. In this review, we compare human and mouse satellite cells and highlight their similarities and differences. We discuss gaps in our knowledge of human satellite cells, compared with that of mouse satellite cells, and suggest ways in which we may advance studies on human satellite cells, particularly by finding new markers and attempting to re-create the human satellite cell niche in vitro.
Collapse
Affiliation(s)
- Luisa Boldrin
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom.
| | | | | |
Collapse
|
48
|
Wakabayashi M, Ito Y, Hamazaki TS, Okochi H. Efficient Myogenic Differentiation of Murine Dermal Sca-1 (−) Cells via Initial Aggregation Culture. Tissue Eng Part A 2010; 16:3251-9. [DOI: 10.1089/ten.tea.2009.0678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mutsumi Wakabayashi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Medical Biochemistry, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuriko Ito
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tatsuo S. Hamazaki
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hitoshi Okochi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Trensz F, Haroun S, Cloutier A, Richter MV, Grenier G. A muscle resident cell population promotes fibrosis in hindlimb skeletal muscles of mdx mice through the Wnt canonical pathway. Am J Physiol Cell Physiol 2010; 299:C939-47. [PMID: 20810909 DOI: 10.1152/ajpcell.00253.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Previous work has pointed to a role for the Wnt canonical pathway in fibrosis formation in aged skeletal muscles. In the present study, we studied the dystrophic mdx mouse, which displays skeletal muscle fibrosis. Our results indicated that the muscle resident stromal cell (mrSC) population in the muscles of dystrophic mice is higher than in the muscles of age-matched wild-type mice. Wnt3a promoted the proliferation of and collagen expression by cultured mrSCs but arrested the growth of and collagen expression by cultured myoblasts. Injections of Wnt3A in the tibialis anterior muscles of adult wild-type mice significantly enhanced the mrSC population and collagen deposition compared with the contralateral muscles. Conversely, an injection of the Wnt antagonist Dickkof protein (DKK1) into the skeletal muscles of mdx mice significantly reduced collagen deposition. These results suggested that the Wnt canonical pathway expands the population of mrSCs and stimulates their production of collagen as observed during aging and in various myopathies.
Collapse
Affiliation(s)
- Frédéric Trensz
- Research Center on Aging, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | |
Collapse
|
50
|
Westerman KA, Penvose A, Yang Z, Allen PD, Vacanti CA. Adult muscle 'stem' cells can be sustained in culture as free-floating myospheres. Exp Cell Res 2010; 316:1966-76. [PMID: 20381487 PMCID: PMC2878880 DOI: 10.1016/j.yexcr.2010.03.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 02/11/2010] [Accepted: 03/27/2010] [Indexed: 02/07/2023]
Abstract
The effectiveness of cell-based therapy to treat muscle disease has been hampered by difficulties in isolating, maintaining and propagating the stem cells that are needed for treatment. Here we report the isolation of muscle-derived stem cells from both young and old mice and their propagation over extended periods of time in culture as "free-floating" myospheres. Analysis of these sphere-forming cells showed that they express stem cell antigen-1 (Sca-1), beta1 integrin (CD29), Thy-1 (CD90), and CD34, but did not express CD45, CD31, or myogenic markers (Pax7, Myf5, and MyoD). We found that cells derived from myospheres and then grown adherently (MDACs) behaved similar to primary myoblasts, in that these cells expressed myogenic markers and were able to easily form multinucleated myotubes. Unlike the parental myospheres but analogous to primary myoblasts, MDACs expressed Pax7, Myf5, and MyoD, indicating that the parent myosphere cells were a more primitive type of cell. In support of this we demonstrated that myospheres were also able to differentiate into adipogenic and osteogenic cells in culture, as well as being able to contribute to injured muscle in vivo. In summary, we report that primitive adult muscle stem cells can be easily isolated and sustained in culture as myospheres.
Collapse
Affiliation(s)
- Karen A. Westerman
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Ashley Penvose
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Zhong Yang
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Paul D. Allen
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Charles A. Vacanti
- Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|