1
|
Yu H, Fan J, Zhang Y, Zhao Z, Lin Z, Jiang P. Syndecan-3 inhibits LPS-induced Inflammation of Bovine Mammary Epithelial Cells through the NF-κB Signal Transduction Pathway. J Dairy Sci 2024:S0022-0302(24)01164-0. [PMID: 39343222 DOI: 10.3168/jds.2024-25212] [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: 05/23/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024]
Abstract
In mastitis, excessive inflammation caused by lipopolysaccharide (LPS) is an important factor leading to mammary tissue damage. Therefore, exploring the regulatory factors that can inhibit the widespread inflammation caused by LPS is crucial. Syndecan-3 (SDC3) has been found to play an active role in anti-inflammatory infection by inhibiting leukocyte adhesion, reducing the accumulation of inflammatory products, such as reactive oxygen species, and competing with chemokines; however, the role and regulatory mechanism of SDC3 in mastitis remains unknown. Therefore, this study aimed to reveal the effect of SDC3 on LPS-induced inflammation in bovine mammary epithelial cells (BMECs) and explore its possible molecular mechanisms. First, we constructed a BMEC inflammatory model. It was found that cells stimulated with 10 μg/mL LPS for 24 h strongly induced the expression of inflammatory cytokines and had no toxic effect on cells, which was the best condition to simulate the BMECs inflammatory response in vitro. Subsequently, we used overexpression and RNAi interference, Real Time Quantitative PCR (RT-qPCR), and Western blot assays to explore the effects of SDC3 on LPS-induced inflammatory factors and their mechanisms. The results showed that overexpression of SDC3 could inhibit the transcriptional levels of inflammatory cytokines IL-6, IL-1β, and TNFα induced by LPS and inhibit the activation of the NF-κB inflammatory pathway by inhibiting the expression of NF-κB p50 and p-IκBα and promoting the expression of IκBα. Our results suggest that SDC3 inhibits the LPS-induced inflammatory response of BMECs through the NF-κB pathway, in which NF-κB p50 may be an important target of SDC3. These findings lay the foundation for elucidating the molecular regulatory mechanisms of dairy cow mastitis.
Collapse
Affiliation(s)
- Haibin Yu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Jing Fan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Yongliang Zhang
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Zhihui Zhao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Ziwei Lin
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China.
| | - Ping Jiang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China.
| |
Collapse
|
2
|
Byun WS, Lee J, Baek JH. Beyond the bulk: overview and novel insights into the dynamics of muscle satellite cells during muscle regeneration. Inflamm Regen 2024; 44:39. [PMID: 39327631 PMCID: PMC11426090 DOI: 10.1186/s41232-024-00354-1] [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: 04/03/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Skeletal muscle possesses remarkable regenerative capabilities, fully recovering within a month following severe acute damage. Central to this process are muscle satellite cells (MuSCs), a resident population of somatic stem cells capable of self-renewal and differentiation. Despite the highly predictable course of muscle regeneration, evaluating this process has been challenging due to the heterogeneous nature of myogenic precursors and the limited insight provided by traditional markers with overlapping expression patterns. Notably, recent advancements in single-cell technologies, such as single-cell (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq), have revolutionized muscle research. These approaches allow for comprehensive profiling of individual cells, unveiling dynamic heterogeneity among myogenic precursors and their contributions to regeneration. Through single-cell transcriptome analyses, researchers gain valuable insights into cellular diversity and functional dynamics of MuSCs post-injury. This review aims to consolidate classical and new insights into the heterogeneity of myogenic precursors, including the latest discoveries from novel single-cell technologies.
Collapse
Affiliation(s)
- Woo Seok Byun
- School of Life Science, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Jinu Lee
- School of Life Science, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea
| | - Jea-Hyun Baek
- School of Life Science, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea.
| |
Collapse
|
3
|
Majchrzak K, Hentschel E, Hönzke K, Geithe C, von Maltzahn J. We need to talk-how muscle stem cells communicate. Front Cell Dev Biol 2024; 12:1378548. [PMID: 39050890 PMCID: PMC11266305 DOI: 10.3389/fcell.2024.1378548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Skeletal muscle is one of the tissues with the highest ability to regenerate, a finely controlled process which is critically depending on muscle stem cells. Muscle stem cell functionality depends on intrinsic signaling pathways and interaction with their immediate niche. Upon injury quiescent muscle stem cells get activated, proliferate and fuse to form new myofibers, a process involving the interaction of multiple cell types in regenerating skeletal muscle. Receptors in muscle stem cells receive the respective signals through direct cell-cell interaction, signaling via secreted factors or cell-matrix interactions thereby regulating responses of muscle stem cells to external stimuli. Here, we discuss how muscle stem cells interact with their immediate niche focusing on how this controls their quiescence, activation and self-renewal and how these processes are altered in age and disease.
Collapse
Affiliation(s)
- Karolina Majchrzak
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Erik Hentschel
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Katja Hönzke
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christiane Geithe
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Julia von Maltzahn
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Faculty for Environment and Natural Sciences, Brandenburg University of Technology Cottbus—Senftenberg, Senftenberg, Germany
| |
Collapse
|
4
|
Rønning SB, Carlsen H, Rocha SDC, Rud I, Solberg N, Høst V, Veiseth-Kent E, Arnesen H, Bergum S, Kirkhus B, Böcker U, Abedali N, Rundblad A, Bålsrud P, Måge I, Holven KB, Ulven SM, Pedersen ME. Dietary intake of micronized avian eggshell membrane in aged mice reduces circulating inflammatory markers, increases microbiota diversity, and attenuates skeletal muscle aging. Front Nutr 2024; 10:1336477. [PMID: 38288061 PMCID: PMC10822908 DOI: 10.3389/fnut.2023.1336477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/27/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Avian eggshell membrane (ESM) is a complex extracellular matrix comprising collagens, glycoproteins, proteoglycans, and hyaluronic acid. We have previously demonstrated that ESM possesses anti-inflammatory properties in vitro and regulates wound healing processes in vivo. The present study aimed to investigate if oral intake of micronized ESM could attenuate skeletal muscle aging associated with beneficial alterations in gut microbiota profile and reduced inflammation. Methods Elderly male C57BL/6 mice were fed an AIN93G diet supplemented with 0, 0.1, 1, or 8% ESM. Young mice were used as reference. The digestibility of ESM was investigated using the static in vitro digestion model INFOGEST for older people and adults, and the gut microbiota profile was analyzed in mice. In addition, we performed a small-scale pre-clinical human study with healthy home-dwelling elderly (>70 years) who received capsules with a placebo or 500 mg ESM every day for 4 weeks and studied the effect on circulating inflammatory markers. Results and discussion Intake of ESM in elderly mice impacted and attenuated several well-known hallmarks of aging, such as a reduction in the number of skeletal muscle fibers, the appearance of centronucleated fibers, a decrease in type IIa/IIx fiber type proportion, reduced gene expression of satellite cell markers Sdc3 and Pax7 and increased gene expression of the muscle atrophy marker Fbxo32. Similarly, a transition toward the phenotypic characteristics of young mice was observed for several proteins involved in cellular processes and metabolism. The digestibility of ESM was poor, especially for the elderly condition. Furthermore, our experiments showed that mice fed with 8% ESM had increased gut microbiota diversity and altered microbiota composition compared with the other groups. ESM in the diet also lowered the expression of the inflammation marker TNFA in mice and in vitro in THP-1 macrophages. In the human study, intake of ESM capsules significantly reduced the inflammatory marker CRP. Altogether, our results suggest that ESM, a natural extracellular biomaterial, may be attractive as a nutraceutical candidate with a possible effect on skeletal muscle aging possibly through its immunomodulating effect or gut microbiota.
Collapse
Affiliation(s)
| | - Harald Carlsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | | | - Ida Rud
- Nofima AS, Food Division, Ås, Norway
| | | | | | | | - Henriette Arnesen
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | | | | | | | - Nada Abedali
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Amanda Rundblad
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pia Bålsrud
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | - Kirsten Bjørklund Holven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| | - Stine Marie Ulven
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | |
Collapse
|
5
|
Yaghi OK, Hanna BS, Langston PK, Michelson DA, Jayewickreme T, Marin-Rodero M, Benoist C, Mathis D. A discrete 'early-responder' stromal-cell subtype orchestrates immunocyte recruitment to injured tissue. Nat Immunol 2023; 24:2053-2067. [PMID: 37932455 PMCID: PMC10792729 DOI: 10.1038/s41590-023-01669-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/05/2023] [Indexed: 11/08/2023]
Abstract
Following acute injury, stromal cells promote tissue regeneration by a diversity of mechanisms. Time-resolved single-cell RNA sequencing of muscle mesenchymal stromal cells (MmSCs) responding to acute injury identified an 'early-responder' subtype that spiked on day 1 and expressed a notable array of transcripts encoding immunomodulators. IL-1β, TNF-α and oncostatin M each strongly and rapidly induced MmSCs transcribing this immunomodulatory program. Macrophages amplified the program but were not strictly required for its induction. Transfer of the inflammatory MmSC subtype, tagged with a unique surface marker, into healthy hindlimb muscle induced inflammation primarily driven by neutrophils and macrophages. Among the abundant inflammatory transcripts produced by this subtype, Cxcl5 was stroma-specific and highly upregulated with injury. Depletion of this chemokine early after injury revealed a substantial impact on recruitment of neutrophils, a prolongation of inflammation to later times and an effect on tissue regeneration. Mesenchymal stromal cell subtypes expressing a comparable inflammatory program were found in a mouse model of muscular dystrophy and in several other tissues and pathologies in both mice and humans. These 'early-responder' mesenchymal stromal cells, already in place, permit rapid and coordinated mobilization and amplification of critical cell collaborators in response to injury.
Collapse
Affiliation(s)
- Omar K Yaghi
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Bola S Hanna
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Teshika Jayewickreme
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Miguel Marin-Rodero
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Fang L, Kuniya T, Harada Y, Yasuda O, Maeda N, Suzuki Y, Kawaguchi D, Gotoh Y. TIMP3 promotes the maintenance of neural stem-progenitor cells in the mouse subventricular zone. Front Neurosci 2023; 17:1149603. [PMID: 37456993 PMCID: PMC10338847 DOI: 10.3389/fnins.2023.1149603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Adult neural stem cells (NSCs) in the mouse subventricular zone (SVZ) serve as a lifelong reservoir for newborn olfactory bulb neurons. Recent studies have identified a slowly dividing subpopulation of embryonic neural stem-progenitor cells (NPCs) as the embryonic origin of adult NSCs. Yet, little is known about how these slowly dividing embryonic NPCs are maintained until adulthood while other NPCs are extinguished by the completion of brain development. The extracellular matrix (ECM) is an essential component of stem cell niches and thus a key determinant of stem cell fate. Here we investigated tissue inhibitors of metalloproteinases (TIMPs)-regulators of ECM remodeling-for their potential roles in the establishment of adult NSCs. We found that Timp2, Timp3, and Timp4 were expressed at high levels in slowly dividing NPCs compared to rapidly dividing NPCs. Deletion of TIMP3 reduced the number of adult NSCs and neuroblasts in the lateral SVZ. In addition, overexpression of TIMP3 in the embryonic NPCs suppressed neuronal differentiation and upregulated the expression levels of Notch signaling relating genes. These results thus suggest that TIMP3 keeps the undifferentiated state of embryonic NPCs, leading to the establishment and maintenance of adult NSCs.
Collapse
Affiliation(s)
- Lingyan Fang
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takaaki Kuniya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yujin Harada
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Osamu Yasuda
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, Kanoya, Japan
| | - Nobuyo Maeda
- Department of Sports and Life Sciences, National Institute of Fitness and Sports in Kanoya, Kanoya, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Daichi Kawaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Ricard-Blum S, Couchman JR. Conformations, interactions and functions of intrinsically disordered syndecans. Biochem Soc Trans 2023:BST20221085. [PMID: 37334846 DOI: 10.1042/bst20221085] [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: 02/19/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans present on most mammalian cell surfaces. They have a long evolutionary history, a single syndecan gene being expressed in bilaterian invertebrates. Syndecans have attracted interest because of their potential roles in development and disease, including vascular diseases, inflammation and various cancers. Recent structural data is providing important insights into their functions, which are complex, involving both intrinsic signaling through cytoplasmic binding partners and co-operative mechanisms where syndecans form a signaling nexus with other receptors such as integrins and tyrosine kinase growth factor receptors. While the cytoplasmic domain of syndecan-4 has a well-defined dimeric structure, the syndecan ectodomains are intrinsically disordered, which is linked to a capacity to interact with multiple partners. However, it remains to fully establish the impact of glycanation and partner proteins on syndecan core protein conformations. Genetic models indicate that a conserved property of syndecans links the cytoskeleton to calcium channels of the transient receptor potential class, compatible with roles as mechanosensors. In turn, syndecans influence actin cytoskeleton organization to impact motility, adhesion and the extracellular matrix environment. Syndecan clustering with other cell surface receptors into signaling microdomains has relevance to tissue differentiation in development, for example in stem cells, but also in disease where syndecan expression can be markedly up-regulated. Since syndecans have potential as diagnostic and prognostic markers as well as possible targets in some forms of cancer, it remains important to unravel structure/function relationships in the four mammalian syndecans.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- ICBMS, UMR 5246 CNRS, Universite Claude Bernard Lyon 1, F-69622 Villeurbanne, France
| | - John R Couchman
- Biotech Research & Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
8
|
Fan J, Zhao Z, Wu H, Fang X, Miao F, Chen X, Jiang X, Li J, Jiang P, Yu H. Syndecan-3 Coregulates Milk Fat Metabolism and Inflammatory Reactions in Bovine Mammary Epithelial Cells through AMPK/SIRT1 Signaling Pathway. Int J Mol Sci 2023; 24:6657. [PMID: 37047630 PMCID: PMC10095454 DOI: 10.3390/ijms24076657] [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: 01/25/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Transcriptome sequencing showed that syndecan-3 (SDC3) was differentially expressed in high-fat and low-fat mammary epithelial cells of Chinese Holstein cows. Previous studies found that SDC3 plays an important role in inflammatory diseases and virus infection. However, those studies did not confirm whether or not the functional gene SDC3, which plays an important role in regulating milk fat metabolism, has an effect on susceptibility to breast tissue diseases. Therefore, we studied the effects of SDC3 on milk lipid metabolism and inflammation in bovine mammary epithelial cells (BMECs) and further explored the common regulatory pathway of SDC3 in both. The overexpression of SDC3 increased the contents of triglycerides and cholesterol, reduced the content of non-esterified fatty acids, inhibited the expression of inflammatory factors (IL-6, IL-1β, TNF-α and COX-2), and reduced the production of ROS in BMECs. However, silenced SDC3 had the opposite effect. Further exploring the mechanisms of SDC3, we found that SDC3 upregulated the expression of peroxisome proliferator-activated receptor gamma (PPARG) through the AMPK/SIRT1 signal pathway to promote milk fat synthesis. It also regulated the activation of the NF-κB pathway through the AMPK/SIRT1 signal pathway, reducing the expression of inflammatory factors and ROS production, thus inhibiting the inflammatory response of BMECs. Nuclear factor kappa B subunit 1 (NF-κB p50) was an important target of SDC3 in this process. To sum up, our results showed that SDC3 coregulated milk fat metabolism and inflammation through the AMPK/SIRT1 signaling pathway. This study laid a foundation for the comprehensive evaluation of breeding value based on multi-effect functional genes in dairy cow molecular breeding.
Collapse
Affiliation(s)
- Jing Fan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Zhihui Zhao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Haochen Wu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Xibi Fang
- College of Animal Science, Jilin University, Changchun 130062, China;
| | - Fengshuai Miao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Xuanxu Chen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Xinyi Jiang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Jing Li
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Ping Jiang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| | - Haibin Yu
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.F.); (Z.Z.); (H.W.); (F.M.); (X.C.); (X.J.); (J.L.)
- The Key Laboratory of Animal Resources and Breed Innovation in Western Guangdong Province, Zhanjiang 524088, China
| |
Collapse
|
9
|
Maciej-Hulme ML, Melrose J, Farrugia BL. Arthritis and Duchenne muscular dystrophy: the role of chondroitin sulfate and its associated proteoglycans in disease pathology and as a diagnostic marker. Am J Physiol Cell Physiol 2023; 324:C142-C152. [PMID: 36409173 PMCID: PMC9829464 DOI: 10.1152/ajpcell.00103.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022]
Abstract
Chondroitin sulfate (CS) is a ubiquitous glycosaminoglycan covalently attached to the core proteins of cell surface, extracellular, and intracellular proteoglycans. The multistep and highly regulated biosynthesis of chondroitin sulfate and its degradation products give rise to a diverse species of molecules with functional regulatory properties in biological systems. This review will elucidate and expand on the most recent advances in understanding the role of chondroitin sulfate and its associate proteoglycans, in arthritis and Duchenne muscular dystrophy (DMD), two different and discrete pathologies. Highlighting not only the biodiverse nature of this family of molecules but also the utilization of CS proteoglycans, CS, and its catabolic fragments as biomarkers and potential therapeutic targets for disease pathologies.
Collapse
Affiliation(s)
- Marissa L Maciej-Hulme
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and The Faculty of Medicine and Health, The University of Sydney, St. Leonard's, New South Wales, Australia
| | - Brooke L Farrugia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Colin-Pierre C, El Baraka O, Danoux L, Bardey V, André V, Ramont L, Brézillon S. Regulation of stem cell fate by HSPGs: implication in hair follicle cycling. NPJ Regen Med 2022; 7:77. [PMID: 36577752 PMCID: PMC9797564 DOI: 10.1038/s41536-022-00267-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/30/2022] [Indexed: 12/29/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are part of proteoglycan family. They are composed of heparan sulfate (HS)-type glycosaminoglycan (GAG) chains covalently linked to a core protein. By interacting with growth factors and/or receptors, they regulate numerous pathways including Wnt, hedgehog (Hh), bone morphogenic protein (BMP) and fibroblast growth factor (FGF) pathways. They act as inhibitor or activator of these pathways to modulate embryonic and adult stem cell fate during organ morphogenesis, regeneration and homeostasis. This review summarizes the knowledge on HSPG structure and classification and explores several signaling pathways regulated by HSPGs in stem cell fate. A specific focus on hair follicle stem cell fate and the possibility to target HSPGs in order to tackle hair loss are discussed in more dermatological and cosmeceutical perspectives.
Collapse
Affiliation(s)
- Charlie Colin-Pierre
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.
- BASF Beauty Care Solutions France SAS, Pulnoy, France.
| | | | - Louis Danoux
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | | | - Valérie André
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
- CHU de Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| |
Collapse
|
11
|
Schüler SC, Liu Y, Dumontier S, Grandbois M, Le Moal E, Cornelison DDW, Bentzinger CF. Extracellular matrix: Brick and mortar in the skeletal muscle stem cell niche. Front Cell Dev Biol 2022; 10:1056523. [PMID: 36523505 PMCID: PMC9745096 DOI: 10.3389/fcell.2022.1056523] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.
Collapse
Affiliation(s)
- Svenja C. Schüler
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - DDW Cornelison
- Division of Biological Sciences Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - C. Florian Bentzinger
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
12
|
Jones FK, Phillips A, Jones AR, Pisconti A. The INSR/AKT/mTOR pathway regulates the pace of myogenesis in a syndecan-3-dependent manner. Matrix Biol 2022; 113:61-82. [PMID: 36152781 DOI: 10.1016/j.matbio.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 11/25/2022]
Abstract
Muscle stem cells (MuSCs) are indispensable for muscle regeneration. A multitude of extracellular stimuli direct MuSC fate decisions from quiescent progenitors to differentiated myocytes. The activity of these signals is modulated by coreceptors such as syndecan-3 (SDC3). We investigated the global landscape of SDC3-mediated regulation of myogenesis using a phosphoproteomics approach which revealed, with the precision level of individual phosphosites, the large-scale extent of SDC3-mediated regulation of signal transduction in MuSCs. We then focused on INSR/AKT/mTOR as a key pathway regulated by SDC3 during myogenesis and mechanistically dissected SDC3-mediated inhibition of insulin receptor signaling in MuSCs. SDC3 interacts with INSR ultimately limiting signal transduction via AKT/mTOR. Both knockdown of INSR and inhibition of AKT rescue Sdc3-/- MuSC differentiation to wild type levels. Since SDC3 is rapidly downregulated at the onset of differentiation, our study suggests that SDC3 acts a timekeeper to restrain proliferating MuSC response and prevent premature differentiation.
Collapse
Affiliation(s)
- Fiona K Jones
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Alexander Phillips
- School of Electrical Engineering, Electronics and Computer Science, University of Liverpool, Liverpool, UK
| | - Andrew R Jones
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Addolorata Pisconti
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
13
|
The Cell Surface Heparan Sulfate Proteoglycan Syndecan-3 Promotes Ovarian Cancer Pathogenesis. Int J Mol Sci 2022; 23:ijms23105793. [PMID: 35628603 PMCID: PMC9145288 DOI: 10.3390/ijms23105793] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans that integrate signaling at the cell surface. By interacting with cytokines, signaling receptors, proteases, and extracellular matrix proteins, syndecans regulate cell proliferation, metastasis, angiogenesis, and inflammation. We analyzed public gene expression datasets to evaluate the dysregulation and potential prognostic impact of Syndecan-3 in ovarian cancer. Moreover, we performed functional in vitro analysis in syndecan-3-siRNA-treated SKOV3 and CAOV3 ovarian cancer cells. In silico analysis of public gene array datasets revealed that syndecan-3 mRNA expression was significantly increased 5.8-fold in ovarian cancer tissues (n = 744) and 3.4-fold in metastases (n = 44) compared with control tissue (n = 46), as independently confirmed in an RNAseq dataset on ovarian serous cystadenocarcinoma tissue (n = 374, controls: n = 133, 3.5-fold increase tumor vs. normal). Syndecan-3 siRNA knockdown impaired 3D spheroid growth and colony formation as stemness-related readouts in SKOV3 and CAOV3 cells. In SKOV3, but not in CAOV3 cells, syndecan-3 depletion reduced cell viability both under basal conditions and under chemotherapy with cisplatin, or cisplatin and paclitaxel. While analysis of the SIOVDB database did not reveal differences in Syndecan-3 expression between patients, sensitive, resistant or refractory to chemotherapy, KM Plotter analysis of 1435 ovarian cancer patients revealed that high syndecan-3 expression was associated with reduced survival in patients treated with taxol and platin. At the molecular level, a reduction in Stat3 activation and changes in the expression of Wnt and notch signaling constituents were observed. Our study suggests that up-regulation of syndecan-3 promotes the pathogenesis of ovarian cancer by modulating stemness-associated pathways.
Collapse
|
14
|
Hudák A, Letoha A, Vizler C, Letoha T. Syndecan-3 as a Novel Biomarker in Alzheimer's Disease. Int J Mol Sci 2022; 23:3407. [PMID: 35328830 PMCID: PMC8955174 DOI: 10.3390/ijms23063407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/17/2022] Open
Abstract
Early diagnosis of Alzheimer's disease (AD) is of paramount importance in preserving the patient's mental and physical health in a fairly manageable condition for a longer period. Reliable AD detection requires novel biomarkers indicating central nervous system (CNS) degeneration in the periphery. Members of the syndecan family of transmembrane proteoglycans are emerging new targets in inflammatory and neurodegenerative disorders. Reviewing the growing scientific evidence on the involvement of syndecans in the pathomechanism of AD, we analyzed the expression of the neuronal syndecan, syndecan-3 (SDC3), in experimental models of neurodegeneration. Initial in vitro studies showed that prolonged treatment of tumor necrosis factor-alpha (TNF-α) increases SDC3 expression in model neuronal and brain microvascular endothelial cell lines. In vivo studies revealed elevated concentrations of TNF-α in the blood and brain of APPSWE-Tau transgenic mice, along with increased SDC3 concentration in the brain and the liver. Primary brain endothelial cells and peripheral blood monocytes isolated from APPSWE-Tau mice exhibited increased SDC3 expression than wild-type controls. SDC3 expression of blood-derived monocytes showed a positive correlation with amyloid plaque load in the brain, demonstrating that SDC3 on monocytes is a good indicator of amyloid pathology in the brain. Given the well-established role of blood tests, the SDC3 expression of monocytes could serve as a novel biomarker for early AD detection.
Collapse
Affiliation(s)
| | - Annamária Letoha
- Albert Szent-Györgyi Clinical Center, Department of Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary;
| | - Csaba Vizler
- Biological Research Centre, Institute of Biochemistry, H-6726 Szeged, Hungary;
| | | |
Collapse
|
15
|
Ma N, Chen D, Lee JH, Kuri P, Hernandez EB, Kocan J, Mahmood H, Tichy ED, Rompolas P, Mourkioti F. Piezo1 regulates the regenerative capacity of skeletal muscles via orchestration of stem cell morphological states. SCIENCE ADVANCES 2022; 8:eabn0485. [PMID: 35302846 PMCID: PMC8932657 DOI: 10.1126/sciadv.abn0485] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/26/2022] [Indexed: 05/08/2023]
Abstract
Muscle stem cells (MuSCs) are essential for tissue homeostasis and regeneration, but the potential contribution of MuSC morphology to in vivo function remains unknown. Here, we demonstrate that quiescent MuSCs are morphologically heterogeneous and exhibit different patterns of cellular protrusions. We classified quiescent MuSCs into three functionally distinct stem cell states: responsive, intermediate, and sensory. We demonstrate that the shift between different stem cell states promotes regeneration and is regulated by the sensing protein Piezo1. Pharmacological activation of Piezo1 is sufficient to prime MuSCs toward more responsive cells. Piezo1 deletion in MuSCs shifts the distribution toward less responsive cells, mimicking the disease phenotype we find in dystrophic muscles. We further demonstrate that Piezo1 reactivation ameliorates the MuSC morphological and regenerative defects of dystrophic muscles. These findings advance our fundamental understanding of how stem cells respond to injury and identify Piezo1 as a key regulator for adjusting stem cell states essential for regeneration.
Collapse
Affiliation(s)
- Nuoying Ma
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delia Chen
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ji-Hyung Lee
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paola Kuri
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Blake Hernandez
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob Kocan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamd Mahmood
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elisia D. Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Panteleimon Rompolas
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
17
|
Gopal S, Amran A, Elton A, Ng L, Pocock R. A somatic proteoglycan controls Notch-directed germ cell fate. Nat Commun 2021; 12:6708. [PMID: 34795288 PMCID: PMC8602670 DOI: 10.1038/s41467-021-27039-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/02/2021] [Indexed: 11/24/2022] Open
Abstract
Communication between the soma and germline optimizes germ cell fate programs. Notch receptors are key determinants of germ cell fate but how somatic signals direct Notch-dependent germ cell behavior is undefined. Here we demonstrate that SDN-1 (syndecan-1), a somatic transmembrane proteoglycan, controls expression of the GLP-1 (germline proliferation-1) Notch receptor in the Caenorhabditis elegans germline. We find that SDN-1 control of a somatic TRP calcium channel governs calcium-dependent binding of an AP-2 transcription factor (APTF-2) to the glp-1 promoter. Hence, SDN-1 signaling promotes GLP-1 expression and mitotic germ cell fate. Together, these data reveal SDN-1 as a putative communication nexus between the germline and its somatic environment to control germ cell fate decisions.
Collapse
Affiliation(s)
- Sandeep Gopal
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, 3800, Australia.
| | - Aqilah Amran
- grid.1002.30000 0004 1936 7857Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria 3800 Australia
| | - Andre Elton
- grid.1002.30000 0004 1936 7857Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria 3800 Australia
| | - Leelee Ng
- grid.1002.30000 0004 1936 7857Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria 3800 Australia
| | - Roger Pocock
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, 3800, Australia.
| |
Collapse
|
18
|
Syndecan-2 expression enriches for hematopoietic stem cells and regulates stem cell repopulating capacity. Blood 2021; 139:188-204. [PMID: 34767029 DOI: 10.1182/blood.2020010447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 08/27/2021] [Indexed: 11/20/2022] Open
Abstract
The discovery of novel hematopoietic stem cell (HSC) surface markers can enhance understanding of HSC identity and function. We have discovered a population of primitive bone marrow (BM) HSCs distinguished by their expression of the heparan sulfate proteoglycan, Syndecan-2, which serves as both a marker and regulator of HSC function. Syndecan-2 expression was increased 10-fold in CD150+CD48-CD34-c-Kit+Sca-1+Lineage- cells (long-term - HSCs, LT-HSCs) compared to differentiated hematopoietic cells. Isolation of BM cells based solely on Syndecan-2 surface expression produced a 24-fold enrichment for LT-HSCs, 6-fold enrichment for alpha-catulin+c-kit+ HSCs, and yielded HSCs with superior in vivo repopulating capacity compared to CD150+ cells. Competitive repopulation assays revealed the HSC frequency to be 17-fold higher in Syndecan-2+CD34-KSL cells compared to Syndecan-2-CD34-KSL cells and indistinguishable from CD150+CD34-KSL cells. Syndecan-2 expression also identified nearly all repopulating HSCs within the CD150+CD34-KSL population. Mechanistically, Syndecan-2 regulates HSC repopulating capacity through control of expression of Cdkn1c (p57) and HSC quiescence. Loss of Syndecan-2 expression caused increased HSC cell cycle entry, downregulation of Cdkn1c and loss of HSC long-term - repopulating capacity. Syndecan-2 is a novel marker of HSCs which regulates HSC repopulating capacity via control of HSC quiescence.
Collapse
|
19
|
Johnson de Sousa Brito FM, Butcher A, Pisconti A, Poulet B, Prior A, Charlesworth G, Sperinck C, Scotto di Mase M, Liu K, Bou-Gharios G, Jurgen van 't Hof R, Daroszewska A. Syndecan-3 enhances anabolic bone formation through WNT signaling. FASEB J 2021; 35:e21246. [PMID: 33769615 PMCID: PMC8251628 DOI: 10.1096/fj.202002024r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
Osteoporosis is the most common age‐related metabolic bone disorder, which is characterized by low bone mass and deterioration in bone architecture, with a propensity to fragility fractures. The best treatment for osteoporosis relies on stimulation of osteoblasts to form new bone and restore bone structure, however, anabolic therapeutics are few and their use is time restricted. Here, we report that Syndecan‐3 increases new bone formation through enhancement of WNT signaling in osteoblasts. Young adult Sdc3−/− mice have low bone volume, reduced bone formation, increased bone marrow adipose tissue, increased bone fragility, and a blunted anabolic bone formation response to mechanical loading. This premature osteoporosis‐like phenotype of Sdc3−/− mice is due to delayed osteoblast maturation and impaired osteoblast function, with contributing increased osteoclast‐mediated bone resorption. Indeed, overexpressing Sdc3 in osteoblasts using the Col1a1 promoter rescues the low bone volume phenotype of the Sdc3−/− mice, and also increases bone volume in WT mice. Mechanistically, SDC3 enhances canonical WNT signaling in osteoblasts through stabilization of Frizzled 1, making SDC3 an attractive target for novel bone anabolic drug development.
Collapse
Affiliation(s)
- Francesca Manuela Johnson de Sousa Brito
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Andrew Butcher
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Addolorata Pisconti
- Department of Biochemistry, IIB, University of Liverpool, Liverpool, UK.,Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Blandine Poulet
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Amanda Prior
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Gemma Charlesworth
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Catherine Sperinck
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Michele Scotto di Mase
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Ke Liu
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - George Bou-Gharios
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Robert Jurgen van 't Hof
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK
| | - Anna Daroszewska
- Department of Musculoskeletal and Ageing Science (formerly Department of Musculoskeletal Biology), Institute of Life Course and Medical Sciences (formerly Institute of Ageing and Chronic Disease), University of Liverpool, Liverpool, UK.,Department of Clinical Biochemistry and Metabolic Medicine, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.,Department of Rheumatology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
20
|
Vilchinskaya NA, Shenkman BS. Myosatellite Cells under Gravitational Unloading Conditions. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
De Micheli AJ, Laurilliard EJ, Heinke CL, Ravichandran H, Fraczek P, Soueid-Baumgarten S, De Vlaminck I, Elemento O, Cosgrove BD. Single-Cell Analysis of the Muscle Stem Cell Hierarchy Identifies Heterotypic Communication Signals Involved in Skeletal Muscle Regeneration. Cell Rep 2021; 30:3583-3595.e5. [PMID: 32160558 DOI: 10.1016/j.celrep.2020.02.067] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/12/2019] [Accepted: 02/14/2020] [Indexed: 12/15/2022] Open
Abstract
Muscle regeneration relies on the regulation of muscle stem cells (MuSCs) through paracrine signaling interactions. We analyzed muscle regeneration in mice using single-cell RNA sequencing (scRNA-seq) and generated over 34,000 single-cell transcriptomes spanning four time-points. We identified 15 distinct cell types including heterogenous populations of muscle stem and progenitor cells. We resolved a hierarchical map of these myogenic cells by trajectory inference and observed stage-specific regulatory programs within this continuum. Through ligand-receptor interaction analysis, we identified over 100 candidate regeneration-associated paracrine communication pairs between MuSCs and non-myogenic cells. We show that myogenic stem/progenitor cells exhibit heterogeneous expression of multiple Syndecan proteins in cycling myogenic cells, suggesting that Syndecans may coordinate myogenic fate regulation. We performed ligand stimulation in vitro and confirmed that three paracrine factors (FGF2, TGFβ1, and RSPO3) regulate myogenic cell proliferation in a Syndecan-dependent manner. Our study provides a scRNA-seq reference resource to investigate cell communication interactions in muscle regeneration.
Collapse
Affiliation(s)
- Andrea J De Micheli
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Emily J Laurilliard
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Charles L Heinke
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Hiranmayi Ravichandran
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Paula Fraczek
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
22
|
Gopal S, Arokiasamy S, Pataki C, Whiteford JR, Couchman JR. Syndecan receptors: pericellular regulators in development and inflammatory disease. Open Biol 2021; 11:200377. [PMID: 33561383 PMCID: PMC8061687 DOI: 10.1098/rsob.200377] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023] Open
Abstract
The syndecans are the major family of transmembrane proteoglycans, usually bearing multiple heparan sulfate chains. They are present on virtually all nucleated cells of vertebrates and are also present in invertebrates, indicative of a long evolutionary history. Genetic models in both vertebrates and invertebrates have shown that syndecans link to the actin cytoskeleton and can fine-tune cell adhesion, migration, junction formation, polarity and differentiation. Although often associated as co-receptors with other classes of receptors (e.g. integrins, growth factor and morphogen receptors), syndecans can nonetheless signal to the cytoplasm in discrete ways. Syndecan expression levels are upregulated in development, tissue repair and an array of human diseases, which has led to the increased appreciation that they may be important in pathogenesis not only as diagnostic or prognostic agents, but also as potential targets. Here, their functions in development and inflammatory diseases are summarized, including their potential roles as conduits for viral pathogen entry into cells.
Collapse
Affiliation(s)
- Sandeep Gopal
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Samantha Arokiasamy
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Csilla Pataki
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - James R. Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - John R. Couchman
- Biotech Research and Innovation Centre, University of Copenhagen, Biocentre 1.3.16, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| |
Collapse
|
23
|
Syndecans in cancer: A review of function, expression, prognostic value, and therapeutic significance. Cancer Treat Res Commun 2021; 27:100312. [PMID: 33485180 DOI: 10.1016/j.ctarc.2021.100312] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
While our understanding of tumors and how to treat them has advanced significantly since the days of Aminopterin and the radical mastectomy, cancer remains among the leading causes of death worldwide. Despite innumerable advancements in medical technology the non-static and highly heterogeneous nature of a tumor can make characterization and treatment exceedingly difficult. Because of this complexity, the identification of new cellular constituents that can be used for diagnostic, prognostic, and therapeutic purposes is crucial in improving patient outcomes worldwide. Growing evidence has demonstrated that among the myriad of changes seen in cancer cells, the Syndecan family of proteins has been observed to undergo drastic alterations in expression. Syndecans are transmembrane heparan sulfate proteoglycans that are responsible for cell signaling, proliferation, and adhesion, and many studies have shed light on their unique involvement in both tumor progression and suppression. This review seeks to discuss Syndecan expression levels in various cancers, whether they make reliable biomarkers for detection and prognosis, and whether they may be viable targets for future cancer therapies. The conclusions drawn from the literature reviewed in this article indicate that changes in expression of Syndecan protein can have profound effects on tumor size, metastatic capability, and overall patient survival rate. Further, while data regarding the therapeutic targeting of Syndecan proteins is sparse, the available literature does demonstrate promise for their use in cancer treatment going forward.
Collapse
|
24
|
Werthmann RC, Tzouros M, Lamerz J, Augustin A, Fritzius T, Trovò L, Stawarski M, Raveh A, Diener C, Fischer C, Gassmann M, Lindemann L, Bettler B. Symmetric signal transduction and negative allosteric modulation of heterodimeric mGlu1/5 receptors. Neuropharmacology 2020; 190:108426. [PMID: 33279506 DOI: 10.1016/j.neuropharm.2020.108426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 11/09/2020] [Accepted: 12/01/2020] [Indexed: 10/22/2022]
Abstract
For a long time metabotropic glutamate receptors (mGluRs) were thought to regulate neuronal functions as obligatory homodimers. Recent reports, however, indicate the existence of heterodimers between group-II and -III mGluRs in the brain, which differ from the homodimers in their signal transduction and sensitivity to negative allosteric modulators (NAMs). Whether the group-I mGluRs, mGlu1 and mGlu5, form functional heterodimers in the brain is still a matter of debate. We now show that mGlu1 and mGlu5 co-purify from brain membranes and hippocampal tissue and co-localize in cultured hippocampal neurons. Complementation assays with mutants deficient in agonist-binding or G protein-coupling reveal that mGlu1/5 heterodimers are functional in heterologous cells and transfected cultured hippocampal neurons. In contrast to heterodimers between group-II and -III mGluRs, mGlu1/5 receptors exhibit a symmetric signal transduction, with both protomers activating G proteins to a similar extent. NAMs of either protomer in mGlu1/5 receptors partially inhibit signaling, showing that both protomers need to be able to reach an active conformation for full receptor activity. Complete heterodimer inhibition is observed when both protomers are locked in their inactive state by a NAM. In summary, our data show that mGlu1/5 heterodimers exhibit a symmetric signal transduction and thus intermediate signaling efficacy and kinetic properties. Our data support the existence of mGlu1/5 heterodimers in neurons and highlight differences in the signaling transduction of heterodimeric mGluRs that influence allosteric modulation.
Collapse
Affiliation(s)
- Ruth C Werthmann
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Manuel Tzouros
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience and Rare Diseases (NRD) (LL, CD, CF), Pharmaceutical Sciences, Biomarkers, Bioinformatics and Omics & Pathology (MT, JL, AA), Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Jens Lamerz
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience and Rare Diseases (NRD) (LL, CD, CF), Pharmaceutical Sciences, Biomarkers, Bioinformatics and Omics & Pathology (MT, JL, AA), Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Angélique Augustin
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience and Rare Diseases (NRD) (LL, CD, CF), Pharmaceutical Sciences, Biomarkers, Bioinformatics and Omics & Pathology (MT, JL, AA), Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Thorsten Fritzius
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Luca Trovò
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Michal Stawarski
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Adi Raveh
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Catherine Diener
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience and Rare Diseases (NRD) (LL, CD, CF), Pharmaceutical Sciences, Biomarkers, Bioinformatics and Omics & Pathology (MT, JL, AA), Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Christophe Fischer
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience and Rare Diseases (NRD) (LL, CD, CF), Pharmaceutical Sciences, Biomarkers, Bioinformatics and Omics & Pathology (MT, JL, AA), Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Lothar Lindemann
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience and Rare Diseases (NRD) (LL, CD, CF), Pharmaceutical Sciences, Biomarkers, Bioinformatics and Omics & Pathology (MT, JL, AA), Roche Innovation Center Basel, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Bernhard Bettler
- Department of Biomedicine, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
25
|
Syndecan-3 regulates MSC adhesion, ERK and AKT signalling in vitro and its deletion enhances MSC efficacy in a model of inflammatory arthritis in vivo. Sci Rep 2020; 10:20487. [PMID: 33235244 PMCID: PMC7686503 DOI: 10.1038/s41598-020-77514-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a debilitating and painful inflammatory autoimmune disease characterised by the accumulation of leukocytes in the synovium, cartilage destruction and bone erosion. The immunomodulatory effects of bone marrow derived mesenchymal stem cells (MSCs) has been widely studied and the recent observations that syndecan-3 (SDC3) is selectively pro-inflammatory in the joint led us to hypothesise that SDC3 might play an important role in MSC biology. MSCs isolated from bone marrow of wild type and Sdc3−/− mice were used to assess immunophenotype, differentiation, adhesion and migration properties and cell signalling pathways. While both cell types show similar differentiation potential and forward scatter values, the cell complexity in wild type MSCs was significantly higher than in Sdc3−/− cells and was accompanied by lower spread surface area. Moreover, Sdc3−/− MSCs adhered more rapidly to collagen type I and showed a dramatic increase in AKT phosphorylation, accompanied by a decrease in ERK1/2 phosphorylation compared with control cells. In a mouse model of antigen-induced inflammatory arthritis, intraarticular injection of Sdc3−/− MSCs yielded enhanced efficacy compared to injection of wild type MSCs. In conclusion, our data suggest that syndecan-3 regulates MSC adhesion and efficacy in inflammatory arthritis, likely via induction of the AKT pathway.
Collapse
|
26
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
27
|
Rønning SB, Carlson CR, Aronsen JM, Pisconti A, Høst V, Lunde M, Liland KH, Sjaastad I, Kolset SO, Christensen G, Pedersen ME. Syndecan-4 -/- Mice Have Smaller Muscle Fibers, Increased Akt/mTOR/S6K1 and Notch/HES-1 Pathways, and Alterations in Extracellular Matrix Components. Front Cell Dev Biol 2020; 8:730. [PMID: 32850844 PMCID: PMC7411008 DOI: 10.3389/fcell.2020.00730] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background Extracellular matrix (ECM) remodeling is essential for skeletal muscle development and adaption in response to environmental cues such as exercise and injury. The cell surface proteoglycan syndecan-4 has been reported to be essential for muscle differentiation, but few molecular mechanisms are known. Syndecan-4–/– mice are unable to regenerate damaged muscle, and display deficient satellite cell activation, proliferation, and differentiation. A reduced myofiber basal lamina has also been reported in syndecan-4–/– muscle, indicating possible defects in ECM production. To get a better understanding of the underlying molecular mechanisms, we have here investigated the effects of syndecan-4 genetic ablation on molecules involved in ECM remodeling and muscle growth, both under steady state conditions and in response to exercise. Methods Tibialis anterior (TA) muscles from sedentary and exercised syndecan-4–/– and WT mice were analyzed by immunohistochemistry, real-time PCR and western blotting. Results Compared to WT, we found that syndecan-4–/– mice had reduced body weight, reduced muscle weight, muscle fibers with a smaller cross-sectional area, and reduced expression of myogenic regulatory transcription factors. Sedentary syndecan-4–/– had also increased mRNA levels of syndecan-2, decorin, collagens, fibromodulin, biglycan, and LOX. Some of these latter ECM components were reduced at protein level, suggesting them to be more susceptible to degradation or less efficiently translated when syndecan-4 is absent. At the protein level, TRPC7 was reduced, whereas activation of the Akt/mTOR/S6K1 and Notch/HES-1 pathways were increased. Finally, although exercise induced upregulation of several of these components in WT, a further upregulation of these molecules was not observed in exercised syndecan-4–/– mice. Conclusion Altogether our data suggest an important role of syndecan-4 in muscle development.
Collapse
Affiliation(s)
| | - Cathrine Rein Carlson
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Bjørknes College, Oslo, Norway
| | - Addolorata Pisconti
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States
| | | | - Marianne Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kristian Hovde Liland
- Nofima AS, Ås, Norway.,Faculty of Sciences and Technology, Norwegian University of Life Sciences, Ås, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | - Svein Olav Kolset
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,K.G. Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| | | |
Collapse
|
28
|
Mingirulli N, Pyle A, Hathazi D, Alston CL, Kohlschmidt N, O'Grady G, Waddell L, Evesson F, Cooper SBT, Turner C, Duff J, Topf A, Yubero D, Jou C, Nascimento A, Ortez C, García‐Cazorla A, Gross C, O'Callaghan M, Santra S, Preece MA, Champion M, Korenev S, Chronopoulou E, Anirban M, Pierre G, McArthur D, Thompson K, Navas P, Ribes A, Tort F, Schlüter A, Pujol A, Montero R, Sarquella G, Lochmüller H, Jiménez‐Mallebrera C, Taylor RW, Artuch R, Kirschner J, Grünert SC, Roos A, Horvath R. Clinical presentation and proteomic signature of patients with TANGO2 mutations. J Inherit Metab Dis 2020; 43:297-308. [PMID: 31339582 PMCID: PMC7078914 DOI: 10.1002/jimd.12156] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
Transport And Golgi Organization protein 2 (TANGO2) deficiency has recently been identified as a rare metabolic disorder with a distinct clinical and biochemical phenotype of recurrent metabolic crises, hypoglycemia, lactic acidosis, rhabdomyolysis, arrhythmias, and encephalopathy with cognitive decline. We report nine subjects from seven independent families, and we studied muscle histology, respiratory chain enzyme activities in skeletal muscle and proteomic signature of fibroblasts. All nine subjects carried autosomal recessive TANGO2 mutations. Two carried the reported deletion of exons 3 to 9, one homozygous, one heterozygous with a 22q11.21 microdeletion inherited in trans. The other subjects carried three novel homozygous (c.262C>T/p.Arg88*; c.220A>C/p.Thr74Pro; c.380+1G>A), and two further novel heterozygous (c.6_9del/p.Phe6del); c.11-13delTCT/p.Phe5del mutations. Immunoblot analysis detected a significant decrease of TANGO2 protein. Muscle histology showed mild variation of fiber diameter, no ragged-red/cytochrome c oxidase-negative fibers and a defect of multiple respiratory chain enzymes and coenzyme Q10 (CoQ10 ) in two cases, suggesting a possible secondary defect of oxidative phosphorylation. Proteomic analysis in fibroblasts revealed significant changes in components of the mitochondrial fatty acid oxidation, plasma membrane, endoplasmic reticulum-Golgi network and secretory pathways. Clinical presentation of TANGO2 mutations is homogeneous and clinically recognizable. The hemizygous mutations in two patients suggest that some mutations leading to allele loss are difficult to detect. A combined defect of the respiratory chain enzymes and CoQ10 with altered levels of several membrane proteins provides molecular insights into the underlying pathophysiology and may guide rational new therapeutic interventions.
Collapse
Affiliation(s)
- Nadja Mingirulli
- Department of Neuropediatrics and Muscle DisordersMedical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
- Department of General PediatricsAdolescent Medicine and Neonatology, Medical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
| | - Angela Pyle
- Wellcome Centre for Mitochondrial ResearchInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Denisa Hathazi
- Biomedical Research DepartmentLeibniz‐Institut für Analytische Wissenschaften – ISAS – e.VDortmundGermany
| | - Charlotte L. Alston
- Wellcome Centre for Mitochondrial ResearchInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUK
| | | | - Gina O'Grady
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Leigh Waddell
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Frances Evesson
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Discipline of Child and Adolescent HealthThe University of SydneySydneyNew South WalesAustralia
| | - Sandra B. T. Cooper
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
- Discipline of Child and Adolescent HealthThe University of SydneySydneyNew South WalesAustralia
| | - Christian Turner
- Discipline of Child and Adolescent HealthThe University of SydneySydneyNew South WalesAustralia
- CardiologyThe Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Jennifer Duff
- Wellcome Centre for Mitochondrial ResearchInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Ana Topf
- John Walton Muscular Dystrophy Research CentreInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
| | - Delia Yubero
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Cristina Jou
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Andrés Nascimento
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Carlos Ortez
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Angels García‐Cazorla
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Claudia Gross
- Wellcome Centre for Mitochondrial ResearchInstitute of Neuroscience, Newcastle UniversityNewcastle upon TyneUK
| | - Maria O'Callaghan
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Saikat Santra
- Birmingham Women's and Children's NHS Foundation TrustBirminghamUK
| | | | | | - Sergei Korenev
- Department of Inherited DiseaseSt Thomas HospitalLondonUK
| | | | - Majumdar Anirban
- South West Regional Metabolic DepartmentBristol Royal Hospital for ChildrenBristolUK
| | - Germaine Pierre
- South West Regional Metabolic DepartmentBristol Royal Hospital for ChildrenBristolUK
| | - Daniel McArthur
- Center for Mendelian Genomics and Program in Medical and Population GeneticsBroad Institute of MIT and HarvardCambridgeMassachusetts
- Analytic and Translational Genetics UnitMassachusetts General HospitalBostonMassachusetts
| | - Kyle Thompson
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Placido Navas
- Centro Andaluz de Biología del DesarrolloUníversidad Pablo de Olavide‐CSIC‐JA and CIBERER, Instituto de Salud Carlos IIIMadridSpain
| | - Antonia Ribes
- Secció d'Errors Congènits del Metabolisme – IBCServei de Bioquímica I Genètìca Molecular, Hospital Clínìc, IDIBAPS, CIBERERBarcelonaSpain
| | - Frederic Tort
- Secció d'Errors Congènits del Metabolisme – IBCServei de Bioquímica I Genètìca Molecular, Hospital Clínìc, IDIBAPS, CIBERERBarcelonaSpain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Institut d'Investìgacío Biomedíca de Bellvitge (IDIBELL), and Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos IIIMadridSpain
| | - Aurora Pujol
- Catalan Institution of Research and Advanced Studies (ICREA)BarcelonaSpain
| | - Raquel Montero
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Georgia Sarquella
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle DisordersMedical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
- Children's Hospital of Eastern Ontario Research Institute, University of OttawaOttawaOntarioCanada
- Division of Neurology, Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - Cecilia Jiménez‐Mallebrera
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Robert W. Taylor
- Kid's Neuroscience Centre, Children's Hospital at WestmeadSydneyNew South WalesAustralia
| | - Rafael Artuch
- Department of Clinical Biochemistry, Genetics, Pediatric Neurology and Cardiology and BiobankInstitut de Recerca Sant Joan de Déu and CIBERER, Instituto de Salud Carlos III BarcelonaBarcelonaSpain
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle DisordersMedical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
| | - Sarah C. Grünert
- Department of General PediatricsAdolescent Medicine and Neonatology, Medical Center – University of Freiburg, Faculty of MedicineBreisgauGermany
| | - Andreas Roos
- Biomedical Research DepartmentLeibniz‐Institut für Analytische Wissenschaften – ISAS – e.VDortmundGermany
- Pediatric NeurologyUniversity Children's Hospital, University of Duisburg‐Essen, Faculty of MedicineEssenGermany
| | - Rita Horvath
- Wellcome Centre for Mitochondrial ResearchInstitute of Genetic Medicine, Newcastle UniversityNewcastle upon TyneUK
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeUK
| |
Collapse
|
29
|
Abstract
The transition between proliferating and quiescent states must be carefully regulated to ensure that cells divide to create the cells an organism needs only at the appropriate time and place. Cyclin-dependent kinases (CDKs) are critical for both transitioning cells from one cell cycle state to the next, and for regulating whether cells are proliferating or quiescent. CDKs are regulated by association with cognate cyclins, activating and inhibitory phosphorylation events, and proteins that bind to them and inhibit their activity. The substrates of these kinases, including the retinoblastoma protein, enforce the changes in cell cycle status. Single cell analysis has clarified that competition among factors that activate and inhibit CDK activity leads to the cell's decision to enter the cell cycle, a decision the cell makes before S phase. Signaling pathways that control the activity of CDKs regulate the transition between quiescence and proliferation in stem cells, including stem cells that generate muscle and neurons. © 2020 American Physiological Society. Compr Physiol 10:317-344, 2020.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA.,Department of Biological Chemistry, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA
| |
Collapse
|
30
|
Abstract
Muscle stem cells, or satellite cells, are required for skeletal muscle maintenance, growth, and repair. Following satellite cell activation, several factors drive asymmetric cell division to generate a stem cell and a proliferative progenitor that forms new muscle. The balance between symmetric self-renewal and asymmetric division significantly impacts the efficiency of regeneration. In this Review, we discuss the relationship of satellite cell heterogeneity and the establishment of polarity to asymmetric division, as well as how these processes are impacted in homeostasis, aging, and disease. We also highlight therapeutic opportunities for targeting satellite cell polarity and self-renewal to stimulate muscle regeneration.
Collapse
|
31
|
Schmidt M, Schüler SC, Hüttner SS, von Eyss B, von Maltzahn J. Adult stem cells at work: regenerating skeletal muscle. Cell Mol Life Sci 2019; 76:2559-2570. [PMID: 30976839 PMCID: PMC6586695 DOI: 10.1007/s00018-019-03093-6] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/16/2022]
Abstract
Skeletal muscle regeneration is a finely tuned process involving the activation of various cellular and molecular processes. Satellite cells, the stem cells of skeletal muscle, are indispensable for skeletal muscle regeneration. Their functionality is critically modulated by intrinsic signaling pathways as well as by interactions with the stem cell niche. Here, we discuss the properties of satellite cells, including heterogeneity regarding gene expression and/or their phenotypic traits and the contribution of satellite cells to skeletal muscle regeneration. We also summarize the process of regeneration with a specific emphasis on signaling pathways, cytoskeletal rearrangements, the importance of miRNAs, and the contribution of non-satellite cells such as immune cells, fibro-adipogenic progenitor cells, and PW1-positive/Pax7-negative interstitial cells.
Collapse
Affiliation(s)
- Manuel Schmidt
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Svenja C Schüler
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Sören S Hüttner
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Björn von Eyss
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany
| | - Julia von Maltzahn
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745, Jena, Germany.
| |
Collapse
|
32
|
Thorsvik S, van Beelen Granlund A, Svendsen TD, Bakke I, Røyset ES, Flo TH, Damås JK, Østvik AE, Bruland T, Sandvik AK. Ulcer-associated cell lineage expresses genes involved in regeneration and is hallmarked by high neutrophil gelatinase-associated lipocalin (NGAL) levels. J Pathol 2019; 248:316-325. [PMID: 30746716 PMCID: PMC6618036 DOI: 10.1002/path.5258] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/13/2019] [Accepted: 02/07/2019] [Indexed: 01/16/2023]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL), also known as Lipocalin 2, is an antimicrobial protein, encoded by the gene LCN2, strongly upregulated in inflammatory bowel disease (IBD) and a promising biomarker for IBD. Here we demonstrate that NGAL is highly expressed in all parts of pyloric metaplasia, also known as the ulcer-associated cell lineage (UACL), a metaplastic cell lineage suggested to play a role in wound healing in Crohn's disease (CD). We further show NGAL expression in regenerative intestinal crypts and in undifferentiated patient-derived colonoids. This indicates that NGAL is important in the tissue regeneration process. The remarkable overexpression of NGAL in UACL led us to explore the pathobiology of these cells by transcriptome-wide RNA sequencing. This study is, to our knowledge, the first to characterize the UACL at this level. Biopsies with UACL and inflamed non-UACL epithelium from the terminal ileum of CD patients and epithelium from healthy controls were laser capture microdissected for RNA sequencing. Among the 180 genes differentially expressed between UACL and control epithelium, the ten most-upregulated genes specific for UACL were MUC5AC, PGC, MUC6, MUC5B, LCN2, POU2AF1, MUC1, SDC3, IGFBP5, and SLC7A5. PDX1 was among the most upregulated in both UACL and inflamed non-UACL epithelium. Immunohistochemistry and iDisco 3D visualization was used to characterize UACL histo-morphologically, and to validate protein expression of 11 selected differentially expressed genes. Among these genes, LCN2, NOTCH2, PHLDA1, IGFBP5, SDC3, BPIFB1, and RCN1 have previously not been linked to UACL. Gene expression results were analyzed for functional implications using MetaCore, showing that differentially expressed genes are enriched for genes involved in cell migration and motility, and for biomarkers of gastrointestinal neoplasia. These results support a role for UACL as part of the reepithelialization process during and after destructive intestinal inflammation. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Silje Thorsvik
- Centre of Molecular Inflammation Research, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Gastroenterology and Hepatology, St Olav's University Hospital, Trondheim, Norway
| | - Atle van Beelen Granlund
- Centre of Molecular Inflammation Research, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tarjei D Svendsen
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Clinic of Medicine, St Olav's University Hospital, Trondheim, Norway
| | - Elin S Røyset
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pathology, St Olav's University Hospital, Trondheim, Norway
| | - Trude H Flo
- Centre of Molecular Inflammation Research, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jan K Damås
- Centre of Molecular Inflammation Research, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Infectious Diseases, St Olav's University Hospital, Trondheim, Norway
| | - Ann E Østvik
- Centre of Molecular Inflammation Research, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Gastroenterology and Hepatology, St Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Clinic of Medicine, St Olav's University Hospital, Trondheim, Norway
| | - Arne K Sandvik
- Centre of Molecular Inflammation Research, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Gastroenterology and Hepatology, St Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
33
|
Karamanos NK, Piperigkou Z, Theocharis AD, Watanabe H, Franchi M, Baud S, Brézillon S, Götte M, Passi A, Vigetti D, Ricard-Blum S, Sanderson RD, Neill T, Iozzo RV. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem Rev 2018; 118:9152-9232. [DOI: 10.1021/acs.chemrev.8b00354] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras 26110, Greece
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi 480-1195, Japan
| | - Marco Franchi
- Department for Life Quality Studies, University of Bologna, Rimini 47100, Italy
| | - Stéphanie Baud
- Université de Reims Champagne-Ardenne, Laboratoire SiRMa, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, Laboratoire de Biochimie Médicale et Biologie Moléculaire, CNRS UMR MEDyC 7369, Faculté de Médecine, 51 rue Cognacq Jay, Reims 51100, France
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster 48149, Germany
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese 21100, Italy
| | - Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, CNRS, UMR 5246, Institute of Molecular and Supramolecular Chemistry and Biochemistry, Villeurbanne 69622, France
| | - Ralph D. Sanderson
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 10107, United States
| |
Collapse
|
34
|
Abreu P. Bioenergetics mechanisms regulating muscle stem cell self-renewal commitment and function. Biomed Pharmacother 2018; 103:463-472. [DOI: 10.1016/j.biopha.2018.04.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 11/25/2022] Open
|
35
|
|
36
|
Flamini V, Ghadiali RS, Antczak P, Rothwell A, Turnbull JE, Pisconti A. The Satellite Cell Niche Regulates the Balance between Myoblast Differentiation and Self-Renewal via p53. Stem Cell Reports 2018; 10:970-983. [PMID: 29429962 PMCID: PMC5918193 DOI: 10.1016/j.stemcr.2018.01.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/18/2022] Open
Abstract
Satellite cells are adult muscle stem cells residing in a specialized niche that regulates their homeostasis. How niche-generated signals integrate to regulate gene expression in satellite cell-derived myoblasts is poorly understood. We undertook an unbiased approach to study the effect of the satellite cell niche on satellite cell-derived myoblast transcriptional regulation and identified the tumor suppressor p53 as a key player in the regulation of myoblast quiescence. After activation and proliferation, a subpopulation of myoblasts cultured in the presence of the niche upregulates p53 and fails to differentiate. When satellite cell self-renewal is modeled ex vivo in a reserve cell assay, myoblasts treated with Nutlin-3, which increases p53 levels in the cell, fail to differentiate and instead become quiescent. Since both these Nutlin-3 effects are rescued by small interfering RNA-mediated p53 knockdown, we conclude that a tight control of p53 levels in myoblasts regulates the balance between differentiation and return to quiescence.
Collapse
Affiliation(s)
- Valentina Flamini
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Rachel S Ghadiali
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Philipp Antczak
- Department of Functional Genomics, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; Computational Biology Facility, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Amy Rothwell
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jeremy E Turnbull
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Addolorata Pisconti
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK.
| |
Collapse
|
37
|
Baghdadi MB, Tajbakhsh S. Regulation and phylogeny of skeletal muscle regeneration. Dev Biol 2018; 433:200-209. [DOI: 10.1016/j.ydbio.2017.07.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/30/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
38
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
39
|
Ghadiali RS, Guimond SE, Turnbull JE, Pisconti A. Dynamic changes in heparan sulfate during muscle differentiation and ageing regulate myoblast cell fate and FGF2 signalling. Matrix Biol 2017; 59:54-68. [PMID: 27496348 PMCID: PMC5380652 DOI: 10.1016/j.matbio.2016.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 01/16/2023]
Abstract
Satellite cells (SCs) are skeletal muscle stem cells residing quiescent around healthy muscle fibres. In response to injury or disease SCs activate, proliferate and eventually differentiate and fuse to one another to form new muscle fibres, or to existing damaged fibres to repair them. The sulfated polysaccharide heparan sulfate (HS) is a highly variable biomolecule known to play key roles in the regulation of cell fate decisions, though the changes that muscle HS undergoes during SC differentiation are unknown. Here we show that the sulfation levels of HS increase during SC differentiation; more specifically, we observe an increase in 6-O and 2-O-sulfation in N-acetylated disaccharides. Interestingly, a specific increase in 6-O sulfation is also observed in the heparanome of ageing muscle, which we show leads to promotion of FGF2 signalling and satellite cell proliferation, suggesting a role for the heparanome dynamics in age-associated loss of quiescence. Addition of HS mimetics to differentiating SC cultures results in differential effects: an oversulfated HS mimetic increases differentiation and inhibits FGF2 signalling, a known major promoter of SC proliferation and inhibitor of differentiation. In contrast, FGF2 signalling is promoted by an N-acetylated HS mimetic, which inhibits differentiation and promotes SC expansion. We conclude that the heparanome of SCs is dynamically regulated during muscle differentiation and ageing, and that such changes might account for some of the phenotypes and signalling events that are associated with these processes.
Collapse
Affiliation(s)
- R S Ghadiali
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - S E Guimond
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - J E Turnbull
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | - A Pisconti
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom.
| |
Collapse
|
40
|
Soriano-Arroquia A, Clegg PD, Molloy AP, Goljanek-Whysall K. Preparation and Culture of Myogenic Precursor Cells/Primary Myoblasts from Skeletal Muscle of Adult and Aged Humans. J Vis Exp 2017. [PMID: 28287512 PMCID: PMC5408649 DOI: 10.3791/55047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Skeletal muscle homeostasis depends on muscle growth (hypertrophy), atrophy and regeneration. During ageing and in several diseases, muscle wasting occurs. Loss of muscle mass and function is associated with muscle fiber type atrophy, fiber type switching, defective muscle regeneration associated with dysfunction of satellite cells, muscle stem cells, and other pathophysiological processes. These changes are associated with changes in intracellular as well as local and systemic niches. In addition to most commonly used rodent models of muscle ageing, there is a need to study muscle homeostasis and wasting using human models, which due to ethical implications, consist predominantly of in vitro cultures. Despite the wide use of human Myogenic Progenitor Cells (MPCs) and primary myoblasts in myogenesis, there is limited data on using human primary myoblast and myotube cultures to study molecular mechanisms regulating different aspects of age-associated muscle wasting, aiding in the validation of mechanisms of ageing proposed in rodent muscle. The use of human MPCs, primary myoblasts and myotubes isolated from adult and aged people, provides a physiologically relevant model of molecular mechanisms of processes associated with muscle growth, atrophy and regeneration. Here we describe in detail a robust, inexpensive, reproducible and efficient protocol for the isolation and maintenance of human MPCs and their progeny — myoblasts and myotubes from human muscle samples using enzymatic digestion. Furthermore, we have determined the passage number at which primary myoblasts from adult and aged people undergo senescence in an in vitro culture. Finally, we show the ability to transfect these myoblasts and the ability to characterize their proliferative and differentiation capacity and propose their suitability for performing functional studies of molecular mechanisms of myogenesis and muscle wasting in vitro.
Collapse
Affiliation(s)
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool
| | - Andrew P Molloy
- Institute of Ageing and Chronic Disease, University of Liverpool; Aintree University Hospital
| | | |
Collapse
|
41
|
Abstract
Skeletal muscle stem cells, originally termed satellite cells for their position adjacent to differentiated muscle fibers, are absolutely required for the process of skeletal muscle repair and regeneration. In the last decade, satellite cells have become one of the most studied adult stem cell systems and have emerged as a standard model not only in the field of stem cell-driven tissue regeneration but also in stem cell dysfunction and aging. Here, we provide background in the field and discuss recent advances in our understanding of muscle stem cell function and dysfunction, particularly in the case of aging, and the potential involvement of muscle stem cells in genetic diseases such as the muscular dystrophies.
Collapse
Affiliation(s)
- Ddw Cornelison
- Division of Biological Sciences and Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| | - 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
|
42
|
Yang W, Yoshida K, Yang B, Huang X. Obstacles and solutions for chemical synthesis of syndecan-3 (53-62) glycopeptides with two heparan sulfate chains. Carbohydr Res 2016; 435:180-194. [PMID: 27810711 PMCID: PMC5110403 DOI: 10.1016/j.carres.2016.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022]
Abstract
Proteoglycans play critical roles in many biological events. Due to their structural complexities, strategies towards synthesis of this class of glycopeptides bearing well-defined glycan chains are urgently needed. In this work, we give the full account of the synthesis of syndecan-3 glycopeptide (53-62) containing two different heparan sulfate chains. For assembly of glycans, a convergent 3+2+3 approach was developed producing two different octasaccharide amino acid cassettes, which were utilized towards syndecan-3 glycopeptides. The glycopeptides presented many obstacles for post-glycosylation manipulation, peptide elongation, and deprotection. Following screening of multiple synthetic sequences, a successful strategy was finally established by constructing partially deprotected single glycan chain containing glycopeptides first, followed by coupling of the glycan-bearing fragments and cleavage of the acyl protecting groups.
Collapse
Affiliation(s)
- Weizhun Yang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI 48824, USA
| | - Keisuke Yoshida
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI 48824, USA
| | - Bo Yang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI 48824, USA.
| |
Collapse
|
43
|
LaFoya B, Munroe JA, Mia MM, Detweiler MA, Crow JJ, Wood T, Roth S, Sharma B, Albig AR. Notch: A multi-functional integrating system of microenvironmental signals. Dev Biol 2016; 418:227-41. [PMID: 27565024 PMCID: PMC5144577 DOI: 10.1016/j.ydbio.2016.08.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/15/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022]
Abstract
The Notch signaling cascade is an evolutionarily ancient system that allows cells to interact with their microenvironmental neighbors through direct cell-cell interactions, thereby directing a variety of developmental processes. Recent research is discovering that Notch signaling is also responsive to a broad variety of stimuli beyond cell-cell interactions, including: ECM composition, crosstalk with other signaling systems, shear stress, hypoxia, and hyperglycemia. Given this emerging understanding of Notch responsiveness to microenvironmental conditions, it appears that the classical view of Notch as a mechanism enabling cell-cell interactions, is only a part of a broader function to integrate microenvironmental cues. In this review, we summarize and discuss published data supporting the idea that the full function of Notch signaling is to serve as an integrator of microenvironmental signals thus allowing cells to sense and respond to a multitude of conditions around them.
Collapse
Affiliation(s)
- Bryce LaFoya
- Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA
| | - Jordan A Munroe
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Masum M Mia
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Michael A Detweiler
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Jacob J Crow
- Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA
| | - Travis Wood
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Steven Roth
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA
| | - Bikram Sharma
- Department of Biological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Allan R Albig
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA; Biomolecular Sciences PhD Program, Boise State University, Boise, ID 83725, USA.
| |
Collapse
|
44
|
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: 29] [Impact Index Per Article: 3.6] [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
|
45
|
Chung H, Multhaupt HAB, Oh ES, Couchman JR. Minireview: Syndecans and their crucial roles during tissue regeneration. FEBS Lett 2016; 590:2408-17. [PMID: 27383370 DOI: 10.1002/1873-3468.12280] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/30/2022]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans, with roles in development, tumorigenesis and inflammation, and growing evidence for involvement in tissue regeneration. This is a fast developing field with the prospect of utilizing tissue engineering and biomaterials in novel therapies. Syndecan receptors are not only ubiquitous in mammalian tissues, regulating cell adhesion, migration, proliferation, and differentiation through independent signaling but also working alongside other receptors. Their importance is highlighted by an ability to interact with a diverse array of ligands, including extracellular matrix glycoproteins, growth factors, morphogens, and cytokines that are important regulators of regeneration. We also discuss the potential for syndecans to regulate stem cell properties, and suggest that understanding these proteoglycans is relevant to exploiting cell, tissue, and materials technologies.
Collapse
Affiliation(s)
- Heesung Chung
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Hinke A B Multhaupt
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Eok-Soo Oh
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - John R Couchman
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| |
Collapse
|
46
|
Pasut A, Chang NC, Gurriaran-Rodriguez U, Faulkes S, Yin H, Lacaria M, Ming H, Rudnicki MA. Notch Signaling Rescues Loss of Satellite Cells Lacking Pax7 and Promotes Brown Adipogenic Differentiation. Cell Rep 2016; 16:333-343. [PMID: 27346341 DOI: 10.1016/j.celrep.2016.06.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 04/22/2016] [Accepted: 05/21/2016] [Indexed: 12/22/2022] Open
Abstract
Pax7 is a nodal transcription factor that is essential for regulating the maintenance, expansion, and myogenic identity of satellite cells during both neonatal and adult myogenesis. Deletion of Pax7 results in loss of satellite cells and impaired muscle regeneration. Here, we show that ectopic expression of the constitutively active intracellular domain of Notch1 (NICD1) rescues the loss of Pax7-deficient satellite cells and restores their proliferative potential. Strikingly NICD1-expressing satellite cells do not undergo myogenic differentiation and instead acquire a brown adipogenic fate both in vivo and in vitro. NICD-expressing Pax7(-/-) satellite cells fail to upregulate MyoD and instead express the brown adipogenic marker PRDM16. Overall, these results show that Notch1 activation compensates for the loss of Pax7 in the quiescent state and acts as a molecular switch to promote brown adipogenesis in adult skeletal muscle.
Collapse
Affiliation(s)
- Alessandra Pasut
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H8M5, Canada
| | - Natasha C Chang
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
| | - Uxia Gurriaran-Rodriguez
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
| | - Sharlene Faulkes
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
| | - Hang Yin
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
| | - Melanie Lacaria
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
| | - Hong Ming
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H8M5, Canada.
| |
Collapse
|
47
|
Arecco N, Clarke CJ, Jones FK, Simpson DM, Mason D, Beynon RJ, Pisconti A. Elastase levels and activity are increased in dystrophic muscle and impair myoblast cell survival, proliferation and differentiation. Sci Rep 2016; 6:24708. [PMID: 27241590 PMCID: PMC4886533 DOI: 10.1038/srep24708] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/04/2016] [Indexed: 01/31/2023] Open
Abstract
In Duchenne muscular dystrophy, progressive loss of muscle tissue is accompanied by fibrosis, chronic inflammation and reduced muscle regenerative capacity. Although much is known about the development of fibrosis and chronic inflammation in muscular dystrophy, less is known about how they are mechanistically linked to loss of muscle regenerative capacity. We have developed a proteomics method to discover dystrophy-associated changes in the muscle progenitor cell niche, which identified serine proteases, and especially neutrophil elastase, as candidates. We show that elastase activity is increased in dystrophic (mdx4cv) muscle and impairs myoblast survival in culture. While the effect of elastase on C2C12 cell survival correlates with the kinetics of elastase-mediated degradation of the substrate to which the cells adhere, the effect of elastase on satellite cell-derived primary myoblast growth and differentiation is substrate-independent and even more dramatic than the effect on C2C12 cells, suggesting a detrimental role for elastase on myogenesis in vivo. Additionally, elastase impairs differentiation of both primary and C2C12 myoblasts into myotubes. Our findings evidence the importance of neutrophil-mediated inflammation in muscular dystrophy and indicate elastase-mediated regulation of myoblast behaviour as a potential mechanism underlying loss of regenerative capacity in dystrophic muscle.
Collapse
Affiliation(s)
- N Arecco
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - C J Clarke
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - F K Jones
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - D M Simpson
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.,Centre for Proteome Research, University of Liverpool, Liverpool L69 7ZB, UK
| | - D Mason
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.,Centre for Cell Imaging, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| | - R J Beynon
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.,Centre for Proteome Research, University of Liverpool, Liverpool L69 7ZB, UK
| | - A Pisconti
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| |
Collapse
|
48
|
Dumont NA, Bentzinger CF, Sincennes MC, Rudnicki MA. Satellite Cells and Skeletal Muscle Regeneration. Compr Physiol 2016; 5:1027-59. [PMID: 26140708 DOI: 10.1002/cphy.c140068] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Skeletal muscles are essential for vital functions such as movement, postural support, breathing, and thermogenesis. Muscle tissue is largely composed of long, postmitotic multinucleated fibers. The life-long maintenance of muscle tissue is mediated by satellite cells, lying in close proximity to the muscle fibers. Muscle satellite cells are a heterogeneous population with a small subset of muscle stem cells, termed satellite stem cells. Under homeostatic conditions all satellite cells are poised for activation by stimuli such as physical trauma or growth signals. After activation, satellite stem cells undergo symmetric divisions to expand their number or asymmetric divisions to give rise to cohorts of committed satellite cells and thus progenitors. Myogenic progenitors proliferate, and eventually differentiate through fusion with each other or to damaged fibers to reconstitute fiber integrity and function. In the recent years, research has begun to unravel the intrinsic and extrinsic mechanisms controlling satellite cell behavior. Nonetheless, an understanding of the complex cellular and molecular interactions of satellite cells with their dynamic microenvironment remains a major challenge, especially in pathological conditions. The goal of this review is to comprehensively summarize the current knowledge on satellite cell characteristics, functions, and behavior in muscle regeneration and in pathological conditions.
Collapse
Affiliation(s)
- Nicolas A Dumont
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - C Florian Bentzinger
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Nestlé Institute of Health Sciences, EPFL Campus, Lausanne, Switzerland
| | - Marie-Claude Sincennes
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael A Rudnicki
- Sprott Centre for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
49
|
|
50
|
Function of Membrane-Associated Proteoglycans in the Regulation of Satellite Cell Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 900:61-95. [DOI: 10.1007/978-3-319-27511-6_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|