1
|
Shim DW, Lee KM, Lee D, Kim JS, Jung YS, Oh SS, Lee SW, Lee JW, Kim BS. Osteochondral Repair with Autologous Cartilage Transplantation with or without Bone Grafting: A Short Pilot Study in Mini-Pigs. Cartilage 2025; 16:61-70. [PMID: 37698092 PMCID: PMC11744595 DOI: 10.1177/19476035231199442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
OBJECTIVE Treatment strategies for osteochondral defects, for which particulated autologous cartilage transplantation (PACT) is an emerging treatment strategy, aim to restore the structure and function of the hyaline cartilage. Herein, we compared the efficacy of PACT with control or human transforming growth factor-β (rhTGF-β), and clarified the necessity of bone graft (BG) with PACT to treat shallow osteochondral defects in a porcine model. DESIGN Two skeletally mature male micropigs received 4 osteochondral defects in each knee. The 16 defects were randomized to (1) empty control, (2) PACT, (3) PACT with BG, or (4) rhTGF-β. Animals were euthanized after 2 months and histomorphometry, immunofluorescence analysis, semiquantitative evaluation (O'Driscoll score), and magnetic resonance observation of cartilage repair tissue (MOCART) score were performed. RESULTS Hyaline cartilages, glycosaminoglycan synthesis, and collagen type II staining were more abundant in the PACT than in the control and rhTGF-β groups. The O'Driscoll score was significantly different between groups (P < 0.001), with both PACT groups showing superiority (P = 0.002). PACT had the highest score (P = 0.002), with improved restoration of subchondral bone compared with PACT with BG. The MOCART score showed significant differences between groups (P = 0.021); MOCART and O'Driscoll scores showed high correlation (r = 0.847, P < 0.001). CONCLUSION Treatment of osteochondral defects with PACT improved tissue quality compared with that with control or rhTGF-β in a porcine model. BG, in addition to PACT, may be unnecessary for shallow osteochondral defects. Clinical Relevance. BG may not be necessary while performing PACT.
Collapse
Affiliation(s)
- Dong Woo Shim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Korea
- Department of Medicine, Inha Graduate School, Incheon, Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Donghyun Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Jun Sik Kim
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Yeon Seop Jung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Sung Suk Oh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Si Wook Lee
- Department of Orthopaedic Surgery, School of Medicine, Dongsan Medical Center, Keimyung University, Daegu, Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Bom Soo Kim
- Department of Orthopaedic Surgery, College of Medicine, Inha University Hospital, Incheon, Korea
| |
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
|
Wu J, Chen J, Ge Y, Huang N, Luo Y. Neuroprotective effect of tanshinone IIA-modified mesenchymal stem cells in a lipopolysaccharide-induced neuroinflammation model. Heliyon 2024; 10:e29424. [PMID: 38638958 PMCID: PMC11024610 DOI: 10.1016/j.heliyon.2024.e29424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
In this study, the neuroprotective potential of tanshinone IIA (TIIA)-modified mesenchymal stem cells (MSC) were investigated using a murine model of lipopolysaccharide (LPS)-induced neuroinflammation. The cognitive performance of the mice was assessed using the Y-maze and Morris water maze tests, while immunofluorescence and Western blot analyses were employed to evaluate the hippocampal expression of pertinent markers and inflammatory factors, respectively. The results from the behavioral experiments demonstrated discernible differences in learning and memory abilities between the model group and the control group (P < 0.05), confirming the successful induction of neuroinflammation. Both the MSC and TIIA-MSC groups exhibited enhancements in the cognitive abilities of neuroinflammatory mice, with the TIIA-MSC group demonstrating a more pronounced improvement (P < 0.01). Immunofluorescence analysis revealed significant activation of microglia in the model group, while the MSC and TIIA-MSC groups exhibited a reduction in hippocampal microglial activation, with the TIIA-MSC group displaying a more substantial decrease. A statistically significant difference in the expression levels of IL-1, IL-6, and TNF-α was observed between the model and control groups (P < 0.05), indicating that IL-1, IL-6, and TNF-α were downregulated in both the MSC and TIIA-MSC groups. Notably, the downregulatory effect was more prominent in the TIIA-MSC group (P < 0.01). Compared to MSC treatment alone, the administration of TIIA-modified MSC demonstrated a superior protective effect against lipopolysaccharide-induced neuroinflammation. These findings underscore the potential therapeutic efficacy of TIIA-modified MSC in mitigating neuroinflammatory responses.
Collapse
Affiliation(s)
- Jingjing Wu
- Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Jian Chen
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Ge
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yong Luo
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
4
|
Rahmati M, Haffner M, Lee MA, Leach JK, Saiz AM. The critical impact of traumatic muscle loss on fracture healing: Basic science and clinical aspects. J Orthop Res 2024; 42:249-258. [PMID: 37990953 DOI: 10.1002/jor.25746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/05/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
Musculoskeletal trauma, specifically fractures, is a leading cause of patient morbidity and disability worldwide. In approximately 20% of cases with fracture and related traumatic muscle loss, bone healing is impaired leading to fracture nonunion. Over the past few years, several studies have demonstrated that bone and the surrounding muscle tissue interact not only anatomically and mechanically but also through biochemical pathways and mediators. Severe damage to the surrounding musculature at the fracture site causes an insufficiency in muscle-derived osteoprogenitor cells that are crucial for fracture healing. As an endocrine tissue, skeletal muscle produces many myokines that act on different bone cells, such as osteoblasts, osteoclasts, osteocytes, and mesenchymal stem cells. Investigating how muscle influences fracture healing at cellular, molecular, and hormonal levels provides translational therapeutic solutions to this clinical challenge. This review provides an overview about the contributions of surrounding muscle tissue in directing fracture healing. The focus of the review is on describing the interactions between bone and muscle in both healthy and fractured environments. We discuss current progress in identifying the bone-muscle molecular pathways and strategies to harness these pathways as cues for accelerating fracture healing. In addition, we review the existing challenges and research opportunities in the field.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| | - Max Haffner
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| | - Mark A Lee
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| | - Jonathan Kent Leach
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
- Department of Biomedical Engineering, University of California, Davis, Davis, California, USA
| | - Augustine M Saiz
- Department of Orthopaedic Surgery, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
5
|
Duo T, Liu X, Mo D, Bian Y, Cai S, Wang M, Li R, Zhu Q, Tong X, Liang Z, Jiang W, Chen S, Chen Y, He Z. Single-base editing in IGF2 improves meat production and intramuscular fat deposition in Liang Guang Small Spotted pigs. J Anim Sci Biotechnol 2023; 14:141. [PMID: 37919760 PMCID: PMC10621156 DOI: 10.1186/s40104-023-00930-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/06/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Chinese indigenous pigs are popular with consumers for their juiciness, flavour and meat quality, but they have lower meat production. Insulin-like growth factor 2 (IGF2) is a maternally imprinted growth factor that promotes skeletal muscle growth by regulating cell proliferation and differentiation. A single nucleotide polymorphism (SNP) within intron 3 of porcine IGF2 disrupts a binding site for the repressor, zinc finger BED-type containing 6 (ZBED6), leading to up-regulation of IGF2 and causing major effects on muscle growth, heart size, and backfat thickness. This favorable mutation is common in Western commercial pig populations, but absent in most Chinese indigenous pig breeds. To improve meat production of Chinese indigenous pigs, we used cytosine base editor 3 (CBE3) to introduce IGF2-intron3-C3071T mutation into porcine embryonic fibroblasts (PEFs) isolated from a male Liang Guang Small Spotted pig (LGSS), and single-cell clones harboring the desired mutation were selected for somatic cell nuclear transfer (SCNT) to generate the founder line of IGF2T/T pigs. RESULTS We found the heterozygous progeny IGF2C/T pigs exhibited enhanced expression of IGF2, increased lean meat by 18%-36%, enlarged loin muscle area by 3%-17%, improved intramuscular fat (IMF) content by 18%-39%, marbling score by 0.75-1, meat color score by 0.53-1.25, and reduced backfat thickness by 5%-16%. The enhanced accumulation of intramuscular fat in IGF2C/T pigs was identified to be regulated by the PI3K-AKT/AMPK pathway, which activated SREBP1 to promote adipogenesis. CONCLUSIONS We demonstrated the introduction of IGF2-intron3-C3071T in Chinese LGSS can improve both meat production and quality, and first identified the regulation of IMF deposition by IGF2 through SREBP1 via the PI3K-AKT/AMPK signaling pathways. Our study provides a further understanding of the biological functions of IGF2 and an example for improving porcine economic traits through precise base editing.
Collapse
Affiliation(s)
- Tianqi Duo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Yu Bian
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Shufang Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
- Hunan Provincial Key Laboratory for Genetic Improvement of Domestic Animal, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Min Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Ruiqiang Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Qi Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Xian Tong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Weilun Jiang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Shiyi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China.
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, Guangdong, China.
| |
Collapse
|
6
|
Tran L, Xie B, Assaf E, Ferrari R, Pipinos II, Casale GP, Alvidrez RIM, Watkins S, Sachdev U. Transcriptomic Profiling Identifies Ferroptosis-Related Gene Signatures in Ischemic Muscle Satellite Cells Affected by Peripheral Artery Disease-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:2023-2029. [PMID: 37675635 PMCID: PMC10549760 DOI: 10.1161/atvbaha.123.319518] [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: 04/26/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND We hypothesized that transcriptomic profiling of muscle satellite cells in peripheral artery disease (PAD) would identify damage-related pathways contributing to skeletal muscle myopathy. We identified a potential role for ferroptosis-a form of programmed lytic cell death by iron-mediated lipid peroxidation-as one such pathway. Ferroptosis promotes myopathy in ischemic cardiac muscle but has an unknown role in PAD. METHODS Muscle satellite cells from donors with PAD were obtained during surgery. cDNA libraries were processed for single-cell RNA sequencing using the 10X Genomics platform. Protein expression was confirmed based on pathways inferred by transcriptomic analysis. RESULTS Unsupervised cluster analysis of over 25 000 cells aggregated from 8 donor samples yielded distinct cell populations grouped by a shared unique transcriptional fingerprint. Quiescent cells were diminished in ischemic muscle while myofibroblasts and apoptotic cells were prominent. Differential gene expression demonstrated a surprising increase in genes associated with iron transport and oxidative stress and a decrease in GPX4 (glutathione peroxidase 4) in ischemic PAD-derived cells. Release of the danger signal HMGB1 (high mobility group box-1) correlated with ferroptotic markers including surface transferrin receptor and were higher in ischemia. Furthermore, lipid peroxidation in muscle satellite cells was modulated by ferrostatin, a ferroptosis inhibitor. Histology confirmed iron deposition and lipofuscin, an inducer of ferroptosis in PAD-affected muscle. CONCLUSIONS This report presents a novel finding that genes known to be involved in ferroptosis are differentially expressed in human skeletal muscle affected by PAD. Targeting ferroptosis may be a novel therapeutic strategy to reduce PAD myopathy.
Collapse
Affiliation(s)
- Lillian Tran
- University of Pittsburgh Medical Center Department of Surgery
| | - Bowen Xie
- University of Pittsburgh Medical Center Department of Surgery
| | - Edwyn Assaf
- University of Pittsburgh Medical Center Department of Surgery
| | - Ricardo Ferrari
- University of Pittsburgh Medical Center Department of Surgery
| | - Iraklis I. Pipinos
- University of Nebraska Medical Center Department of Surgery and the VAResearch Service, VA Nebraska-Western Iowa Health Care System
| | - George P. Casale
- University of Nebraska Medical Center Department of Surgery and the VAResearch Service, VA Nebraska-Western Iowa Health Care System
| | | | - Simon Watkins
- University of Pittsburgh Center for Biologic Imaging
| | - Ulka Sachdev
- University of Pittsburgh Medical Center Department of Surgery
| |
Collapse
|
7
|
Di Conza G, Barbaro F, Zini N, Spaletta G, Remaggi G, Elviri L, Mosca S, Caravelli S, Mosca M, Toni R. Woven bone formation and mineralization by rat mesenchymal stromal cells imply increased expression of the intermediate filament desmin. Front Endocrinol (Lausanne) 2023; 14:1234569. [PMID: 37732119 PMCID: PMC10507407 DOI: 10.3389/fendo.2023.1234569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/07/2023] [Indexed: 09/22/2023] Open
Abstract
Background Disordered and hypomineralized woven bone formation by dysfunctional mesenchymal stromal cells (MSCs) characterize delayed fracture healing and endocrine -metabolic bone disorders like fibrous dysplasia and Paget disease of bone. To shed light on molecular players in osteoblast differentiation, woven bone formation, and mineralization by MSCs we looked at the intermediate filament desmin (DES) during the skeletogenic commitment of rat bone marrow MSCs (rBMSCs), where its bone-related action remains elusive. Results Monolayer cultures of immunophenotypically- and morphologically - characterized, adult male rBMSCs showed co-localization of desmin (DES) with vimentin, F-actin, and runx2 in all cell morphotypes, each contributing to sparse and dense colonies. Proteomic analysis of these cells revealed a topologically-relevant interactome, focused on cytoskeletal and related enzymes//chaperone/signalling molecules linking DES to runx2 and alkaline phosphatase (ALP). Osteogenic differentiation led to mineralized woven bone nodules confined to dense colonies, significantly smaller and more circular with respect to controls. It significantly increased also colony-forming efficiency and the number of DES-immunoreactive dense colonies, and immunostaining of co-localized DES/runx-2 and DES/ALP. These data confirmed pre-osteoblastic and osteoblastic differentiation, woven bone formation, and mineralization, supporting DES as a player in the molecular pathway leading to the osteogenic fate of rBMSCs. Conclusion Immunocytochemical and morphometric studies coupled with proteomic and bioinformatic analysis support the concept that DES may act as an upstream signal for the skeletogenic commitment of rBMSCs. Thus, we suggest that altered metabolism of osteoblasts, woven bone, and mineralization by dysfunctional BMSCs might early be revealed by changes in DES expression//levels. Non-union fractures and endocrine - metabolic bone disorders like fibrous dysplasia and Paget disease of bone might take advantage of this molecular evidence for their early diagnosis and follow-up.
Collapse
Affiliation(s)
- Giusy Di Conza
- Department of Medicine and Surgery - DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine - BIOMED, University of Parma, Parma, Italy
| | - Fulvio Barbaro
- Department of Medicine and Surgery - DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine - BIOMED, University of Parma, Parma, Italy
| | - Nicoletta Zini
- Unit of Bologna, National Research Council of Italy (CNR) Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giulia Spaletta
- Department of Statistical Sciences, University of Bologna, Bologna, Italy
| | - Giulia Remaggi
- Food and Drug Department, University of Parma, Parma, Italy
| | - Lisa Elviri
- Food and Drug Department, University of Parma, Parma, Italy
| | - Salvatore Mosca
- Course on Disorders of the Locomotor System, Fellow Program in Orthopaedics and Traumatology, University Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Caravelli
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Massimiliano Mosca
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Roberto Toni
- Department of Medicine and Surgery - DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine - BIOMED, University of Parma, Parma, Italy
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic, Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies (OSTEONET) Unit, Galliera Medical Center (GMC), San Venanzio di Galliera, BO, Italy
- Section IV - Medical Sciences, Academy of Sciences of the Institute of Bologna, Bologna, Italy
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center - Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
8
|
Tamaki T, Natsume T, Katoh A, Nakajima N, Saito K, Fukuzawa T, Otake M, Enya S, Kangawa A, Imai T, Tamaki M, Uchiyama Y. Differentiation Capacity of Porcine Skeletal Muscle-Derived Stem Cells as Intermediate Species between Mice and Humans. Int J Mol Sci 2023; 24:9862. [PMID: 37373009 DOI: 10.3390/ijms24129862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Large animal experiments are important for preclinical studies of regenerative stem cell transplantation therapy. Therefore, we investigated the differentiation capacity of pig skeletal muscle-derived stem cells (Sk-MSCs) as an intermediate model between mice and humans for nerve muscle regenerative therapy. Enzymatically extracted cells were obtained from green-fluorescence transgenic micro-mini pigs (GFP-Tg MMP) and sorted as CD34+/45- (Sk-34) and CD34-/45-/29+ (Sk-DN) fractions. The ability to differentiate into skeletal muscle, peripheral nerve, and vascular cell lineages was examined via in vitro cell culture and in vivo cell transplantation into the damaged tibialis anterior muscle and sciatic nerves of nude mice and rats. Protein and mRNA levels were analyzed using RT-PCR, immunohistochemistry, and immunoelectron microscopy. The myogenic potential, which was tested by Pax7 and MyoD expression and the formation of muscle fibers, was higher in Sk-DN cells than in Sk-34 cells but remained weak in the latter. In contrast, the capacity to differentiate into peripheral nerve and vascular cell lineages was significantly stronger in Sk-34 cells. In particular, Sk-DN cells did not engraft to the damaged nerve, whereas Sk-34 cells showed active engraftment and differentiation into perineurial/endoneurial cells, endothelial cells, and vascular smooth muscle cells, similar to the human case, as previously reported. Therefore, we concluded that Sk-34 and Sk-DN cells in pigs are closer to those in humans than to those in mice.
Collapse
Affiliation(s)
- Tetsuro Tamaki
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Physiology, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Toshiharu Natsume
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Physiology, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Akira Katoh
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Physiology, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Nobuyuki Nakajima
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Urology, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Kosuke Saito
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Otolaryngology, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Tsuyoshi Fukuzawa
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Radiation Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Masayoshi Otake
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, 2780 Nishikata, Kikugawa 439-0037, Japan
| | - Satoko Enya
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, 2780 Nishikata, Kikugawa 439-0037, Japan
| | - Akihisa Kangawa
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, 2780 Nishikata, Kikugawa 439-0037, Japan
| | - Takeshi Imai
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Miyu Tamaki
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Yoshiyasu Uchiyama
- Muscle Physiology and Cell Biology Unit, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan
| |
Collapse
|
9
|
Pax7 + Satellite Cells in Human Skeletal Muscle After Exercise: A Systematic Review and Meta-analysis. Sports Med 2023; 53:457-480. [PMID: 36266373 DOI: 10.1007/s40279-022-01767-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Skeletal muscle has extraordinary regenerative capabilities against challenge, mainly owing to its resident muscle stem cells, commonly identified by Pax7+, which expediently donate nuclei to the regenerating multinucleated myofibers. This local reserve of stem cells in damaged muscle tissues is replenished by undifferentiated bone marrow stem cells (CD34+) permeating into the surrounding vascular system. OBJECTIVE The purpose of the study was to provide a quantitative estimate for the changes in Pax7+ muscle stem cells (satellite cells) in humans following an acute bout of exercise until 96 h, in temporal relation to circulating CD34+ bone marrow stem cells. A subgroup analysis of age was also performed. METHODS Four databases (Web of Science, PubMed, Scopus, and BASE) were used for the literature search until February 2022. Pax7+ cells in human skeletal muscle were the primary outcome. Circulating CD34+ cells were the secondary outcome. The standardized mean difference (SMD) was calculated using a random-effects meta-analysis. Subgroup analyses were conducted to examine the influence of age, training status, type of exercise, and follow-up time after exercise. RESULTS The final search identified 20 studies for Pax7+ cells comprising a total of 370 participants between the average age of 21 and 74 years and 26 studies for circulating CD34+ bone marrow stem cells comprising 494 participants between the average age of 21 and 67 years. Only one study assessed Pax7+ cells immediately after aerobic exercise and showed a 32% reduction in exercising muscle followed by a fast repletion to pre-exercise level within 3 h. A large effect on increasing Pax7+ cell content in skeletal muscles was observed 24 h after resistance exercise (SMD = 0.89, p < 0.001). Pax7+ cells increased to ~ 50% above pre-exercise level 24-72 h after resistance exercise. For a subgroup analysis of age, a large effect (SMD = 0.81, p < 0.001) was observed on increasing Pax7+ cells in exercised muscle among adults aged > 50 years, whereas adults at younger age presented a medium effect (SMD = 0.64, p < 0.001). Both resistance exercise and aerobic exercise showed a medium overall effect in increasing circulating CD34+ cells (SMD = 0.53, p < 0.001), which declined quickly to the pre-exercise baseline level after exercise within 6 h. CONCLUSIONS An immediate depletion of Pax7+ cells in exercising skeletal muscle concurrent with a transient release of CD34+ cells suggest a replenishment of the local stem cell reserve from bone marrow. A protracted Pax7+ cell expansion in the muscle can be observed during 24-72 h after resistance exercise. This result provides a scientific basis for exercise recommendations on weekly cycles allowing for adequate recovery time. Exercise-induced Pax7+ cell expansion in muscle remains significant at higher age, despite a lower stem cell reserve after age 50 years. More studies are required to confirm whether Pax7+ cell increment can occur after aerobic exercise. CLINICAL TRIAL REGISTRATION Registered at the International Prospective Register of Systematic Reviews (PROSPERO) [identification code CRD42021265457].
Collapse
|
10
|
Lu A, Tseng C, Guo P, Gao Z, Whitney KE, Kolonin MG, Huard J. The role of the aging microenvironment on the fate of PDGFRβ lineage cells in skeletal muscle repair. Stem Cell Res Ther 2022; 13:405. [PMID: 35932084 PMCID: PMC9356493 DOI: 10.1186/s13287-022-03072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/20/2022] [Indexed: 11/20/2022] Open
Abstract
Background During aging, perturbation of muscle progenitor cell (MPC) constituents leads to progressive loss of muscle mass and accumulation of adipose and fibrotic tissue. Mesenchymal stem cells (MSCs) give rise to adipocytes and fibroblasts that accumulate in injured and pathological skeletal muscle through constitutive activation of platelet-derived growth factor receptors (PDGFRs). Although the role of the PDGFRα has been widely explored, there is a paucity of evidence demonstrating the role of PDGFRβ in aged skeletal muscle. Methods In this study, we investigated the role of PDGFRβ lineage cells in skeletal muscle during aging by using Cre/loxP lineage tracing technology. The PDGFR-Cre mice were crossed with global double-fluorescent Cre reporter mice (mTmG) that indelibly marks PDGFRβ lineage cells. Those cells were analyzed and compared at different ages in the skeletal muscle of the mice. Results Our results demonstrated that PDGFRβ lineage cells isolated from the muscles of young mice are MPC-like cells that exhibited satellite cell morphology, expressed Pax7, and undergo myogenic differentiation producing myosin heavy chain expressing myotubes. Conversely, the PDGFRβ lineage cells isolated from muscles of old mice displayed MSC morphology with a reduced myogenic differentiation potential while expressing adipogenic and fibrotic differentiation markers. PDGFRβ lineage cells also gave rise to newly regenerated muscle fibers in young mice after muscle injury, but their muscle regenerative process is reduced in old mice. Conclusions Our data suggest that PDGFRβ lineage cells function as MPCs in young mice, while the same PDGFRβ lineage cells from old mice undergo a fate switch participating in adipose and fibrotic tissue infiltration in aged muscle. The inhibition of fate-switching in PDGFRβ lineage cells may represent a potential approach to prevent fibrosis and fatty infiltration in skeletal muscle during the aging process.
Collapse
Affiliation(s)
- Aiping Lu
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, 181 West Meadow Drive, Suite 1000, Vail, CO, 81657, USA.
| | - Chieh Tseng
- M.D. Anderson Cancer Center, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Ping Guo
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, 181 West Meadow Drive, Suite 1000, Vail, CO, 81657, USA
| | - Zhanguo Gao
- Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Kaitlyn E Whitney
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, 181 West Meadow Drive, Suite 1000, Vail, CO, 81657, USA
| | - Mikhail G Kolonin
- Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, 181 West Meadow Drive, Suite 1000, Vail, CO, 81657, USA.
| |
Collapse
|
11
|
Bone morphogenetic protein 4 rescues the bone regenerative potential of old muscle-derived stem cells via regulation of cell cycle inhibitors. Stem Cell Res Ther 2022; 13:385. [PMID: 35907860 PMCID: PMC9338549 DOI: 10.1186/s13287-022-03047-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bone morphogenetic protein 4 (BMP4) promotes the osteogenic differentiation and the bone regenerative potential of muscle-derived stem cells (MDSCs). BMP4 also promotes the self-renewal of both embryonic and somatic stem cells; however, BMP4 signaling activity significantly decreases with age. Cyclin-dependent kinase inhibitors P16INK4A (P16) and P18INK4C (P18) induce early G1-phase cell cycle blockade by targeting cyclin-dependent kinase 4/6. It is still unclear if BMP4 affects the bone regenerative potential of old MDSCs through regulation of P16 and P18 expression. METHODS Young and old MDSCs were isolated from 3 week (young) and 2-year-old (old) mice. In vitro cell proliferation and multipotent differentiation were performed for young and old MDSCs both before and after BMP4/GFP transduction. Cell cycle genes were analyzed using Q-PCR. The bone regenerative potential of young and old MDSCs transduced with BMP4/GFP were compared using Micro-CT and histological analysis. The bone regenerative potential of young and old MDSCs was also compared between single and double transduction (higher BMP4 levels expression). The cell proliferation, mitochondrial function and osteogenic differentiation was also compared in vitro between cells that have been transduced with BMP4GFP (single and double transduction). The correlation of bone regeneration capacity of young and old MDSCs with P16 and P18 expression was further evaluated at 10 days after cell transplantation using histology and western blot analysis. RESULTS Old murine MDSCs (MDSCs) exhibit reduced proliferation and multi-lineage differentiation potential with or without BMP4 stimulation, when compared to young murine MDSCs. Old MDSCs express significantly higher P16 and lower P18, with more cells in the G0/1 phase and fewer cells in the G2/M phase, compared to young MDSCs. Old MDSCs retrovirally transduced to express BMP4 regenerated less bone in a critical size skull defect in CD-1 nude mice when compared to young retrovirally transduced MDSCs expressing similar BMP4 levels and contribute less to the new regenerated new bone. Importantly, both young and old MDSCs can regenerate more bone when BMP4 expression levels are increased by double-transduction with the retroviral-BMP4/GFP. However, the bone regeneration enhancement with elevated BMP4 was more profound in old MDSCs (400% at 2 weeks) compared to young MDSCs (200%). Accordingly, P18 is upregulated while P16 is downregulated after BMP4 transduction. Double transduction did not further increase cell proliferation nor mitochondrial function but did significantly increase Osx expression in both young and old MDSCs. Old MDSCs had even significant higher Osx levels as compared to young MDSCs following double transduction, while a similar Alp expression was observed between young and old MDSCs after double transduction. In addition, at 10 days after cell transplantation, old MDSCs having undergone double transduction regenerated bone more rapidly as showed by Alcian blue and Von Kossa staining. Western blot assays demonstrated that old MDSCs after retro-BMP4/GFP double transduction have significantly lower P18 expression levels when compared to young BMP4-transduced MDSCs. In addition, P18 expression was slightly increased in old MDSCs after double transduction when compared to single transduction. P16 expression was not detectable for both young and two old BMP4/GFP transduced MDSCs groups. CONCLUSIONS In summary, BMP4 can offset the adverse effect of aging on the osteogenic differentiation and the bone regenerative potential of old MDSCs via up-regulation of P18 and down-regulation P16 expression.
Collapse
|
12
|
Ferrari R, Cong G, Chattopadhyay A, Xie B, Assaf E, Morder K, Calderon MJ, Watkins SC, Sachdev U. Attenuated cell-cycle division protein 2 and elevated mitotic roles of polo-like kinase 1 characterize deficient myoblast fusion in peripheral arterial disease. Biochem Biophys Res Commun 2022; 609:163-168. [PMID: 35436627 PMCID: PMC10687717 DOI: 10.1016/j.bbrc.2022.03.161] [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: 02/27/2022] [Revised: 03/26/2022] [Accepted: 03/31/2022] [Indexed: 11/02/2022]
Abstract
INTRODUCTION We propose that MuSC-derived myoblasts in PAD have transcriptomic differences that can highlight underlying causes of ischemia-induced myopathy. METHODS Differentiation capacity among perfused and ischemic human myoblasts was compared. Following next generation sequencing of mRNA, Ingenuity Pathway Analysis (IPA) was performed for canonical pathway enrichment. Live cell imaging and immunofluorescence were performed to determine myocyte fusion index and protein expression based on insights from IPA, specifically concerning cell cycle regulators including cell-division cycle protein 2 (CDC2) and polo-like kinase 1 (PLK1). RESULTS Ischemic myoblasts formed attenuated myotubes indicative of reduced fusion. Additionally, myoblasts from ischemic segments showed significant differences in canonical pathways associated with PLK1 (upregulated) and G2/M DNA damage checkpoint regulation (downregulated). PLK1 inhibition with BI2536 did not affect cell viability in any group over 24 h but deterred fusion more significantly in PAD myoblasts. Furthermore, PLK1 inhibition reduced the expression of checkpoint protein CDC2 in perfused but not ischemic cells. CONCLUSION Differentiating myoblasts derived from ischemic muscle have significant differences in gene expression including those essential to DNA-damage checkpoint regulation and cell cycle progress. DNA-damage checkpoint dysregulation may contribute to myopathy in PAD.
Collapse
Affiliation(s)
- Ricardo Ferrari
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | - Guangzhi Cong
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA; Department of Cardiology, Cardiovascular Institute, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | | | - B Xie
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | - E Assaf
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | - K Morder
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA
| | | | | | - Ulka Sachdev
- University of Pittsburgh Medical Center Department of Surgery, Division of Vascular Surgery, USA.
| |
Collapse
|
13
|
Lu A, Guo P, Pan H, Tseng C, Sinha KM, Yang F, Scibetta A, Cui Y, Huard M, Zhong L, Ravuri S, Huard J. Enhancement of myogenic potential of muscle progenitor cells and muscle healing during pregnancy. FASEB J 2021; 35:e21378. [PMID: 33565161 DOI: 10.1096/fj.202001914r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/10/2020] [Accepted: 01/04/2021] [Indexed: 11/11/2022]
Abstract
The decline of muscle regenerative potential with age has been attributed to a diminished responsiveness of muscle progenitor cells (MPCs). Heterochronic parabiosis has been used as a model to study the effects of aging on stem cells and their niches. These studies have demonstrated that, by exposing old mice to a young systemic environment, aged progenitor cells can be rejuvenated. One interesting idea is that pregnancy represents a unique biological model of a naturally shared circulatory system between developing and mature organisms. To test this hypothesis, we evaluated the muscle regeneration potential of pregnant mice using a cardiotoxin (CTX) injury mouse model. Our results indicate that the pregnant mice demonstrate accelerated muscle healing compared to nonpregnant control mice following muscle injury based on improved muscle histology, superior muscle regeneration, and a reduction in inflammation and necrosis. Additionally, we found that MPCs isolated from pregnant mice display a significant improvement of myogenic differentiation capacity in vitro and muscle regeneration in vivo when compared to the MPCs from nonpregnant mice. Furthermore, MPCs from nonpregnant mice display enhanced myogenic capacity when cultured in the presence of serum obtained from pregnant mice. Our proteomics data from these studies provides potential therapeutic targets to enhance the myogenic potential of progenitor cells and muscle repair.
Collapse
Affiliation(s)
- Aiping Lu
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Ping Guo
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Haiying Pan
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chieh Tseng
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Krishna M Sinha
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fan Yang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alex Scibetta
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yan Cui
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Ling Zhong
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
14
|
Tuysuz EC, Ozbey U, Gulluoglu S, Kuskucu A, Sahin F, Bayrak OF. miRNAs as cell fate determinants of lateral and paraxial mesoderm differentiation from embryonic stem cells. Dev Biol 2021; 478:212-221. [PMID: 34245726 DOI: 10.1016/j.ydbio.2021.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 10/20/2022]
Abstract
To date, the role of miRNAs on pluripotency and differentiation of ESCs into specific lineages has been studied extensively. However, the specific role of miRNAs during lateral and paraxial mesoderm cell fate decision is still unclear. To address this, we firstly determined miRNA profile of mouse ESCs differentiating towards lateral and paraxial lineages which were detected using Flk1 and PDGFαR antibodies, and of myogenic and hematopoietic differentiation potential of purified paraxial and lateral mesodermal cells within these populations. miRNAs associated with lateral and paraxial mesoderm, and their targets were identified using bioinformatics tools. The targets of the corresponding miRNAs were validated after transfection into mouse ESCs. The roles of the selected miRNAs in lateral, and paraxial mesoderm formation were assessed along with hematopoietic and myogenic differentiation capacity. Among the miRNAs, mmu-miR-126a-3p, mmu-miR-335-5p and mmu-miR-672-5p, upregulated in lateral mesoderm cells, and mmu-miR-10b-5p, mmu-miR-196a-5p and mmu-miR-615-3p, upregulated in paraxial mesoderm cells. While transient co-transfection of mmu-miR-126a-3p, mmu-miR-335-5p and mmu-miR-672-5p increased the number of lateral mesodermal cells, co-transfection of mmu-miR-10b-5p, mmu-miR-196a-5p and mmu-miR-615-3p increased the number of paraxial mesodermal cells. Moreover, differentiation potential of the lateral mesodermal cells into hematopoietic cell lineage increased upon co-transfection of mmu-miR-126a-3p, mmu-miR-335-5p and mmu-miR-672-5p and differentiation potential of the paraxial mesodermal cells into skeletal muscle lineage were increased upon co-transfection of mmu-miR-10b-5p, mmu-miR-196a-5p and mmu-miR-615-3p. In conclusion, we determined the miRNA profile of lateral and paraxial mesodermal cells and co-transfection of miRNAs increased differentiation potential of both lateral and paraxial mesodermal cells transiently.
Collapse
Affiliation(s)
- Emre Can Tuysuz
- Department of Medical Genetics, Yeditepe University Medical School, 34755, Istanbul, Turkey; Department of Genetics and Bioengineering, Yeditepe University, 34755, Istanbul, Turkey
| | - Utku Ozbey
- Department of Medical Genetics, Yeditepe University Medical School, 34755, Istanbul, Turkey; Department of Genetics and Bioengineering, Yeditepe University, 34755, Istanbul, Turkey
| | - Sukru Gulluoglu
- Department of Immunology, School of Medicine, Marmara University, 34854, Istanbul, Turkey
| | - Aysegul Kuskucu
- Department of Medical Genetics, Yeditepe University Medical School, 34755, Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Yeditepe University, 34755, Istanbul, Turkey
| | - Omer Faruk Bayrak
- Department of Medical Genetics, Yeditepe University Medical School, 34755, Istanbul, Turkey.
| |
Collapse
|
15
|
Alarcin E, Bal-Öztürk A, Avci H, Ghorbanpoor H, Dogan Guzel F, Akpek A, Yesiltas G, Canak-Ipek T, Avci-Adali M. Current Strategies for the Regeneration of Skeletal Muscle Tissue. Int J Mol Sci 2021; 22:5929. [PMID: 34072959 PMCID: PMC8198586 DOI: 10.3390/ijms22115929] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic injuries, tumor resections, and degenerative diseases can damage skeletal muscle and lead to functional impairment and severe disability. Skeletal muscle regeneration is a complex process that depends on various cell types, signaling molecules, architectural cues, and physicochemical properties to be successful. To promote muscle repair and regeneration, various strategies for skeletal muscle tissue engineering have been developed in the last decades. However, there is still a high demand for the development of new methods and materials that promote skeletal muscle repair and functional regeneration to bring approaches closer to therapies in the clinic that structurally and functionally repair muscle. The combination of stem cells, biomaterials, and biomolecules is used to induce skeletal muscle regeneration. In this review, we provide an overview of different cell types used to treat skeletal muscle injury, highlight current strategies in biomaterial-based approaches, the importance of topography for the successful creation of functional striated muscle fibers, and discuss novel methods for muscle regeneration and challenges for their future clinical implementation.
Collapse
Affiliation(s)
- Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34854 Istanbul, Turkey;
| | - Ayca Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, 34010 Istanbul, Turkey;
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Hüseyin Avci
- Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Cellular Therapy and Stem Cell Research Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Translational Medicine Research and Clinical Center, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Hamed Ghorbanpoor
- AvciBio Research Group, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey;
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
- Department of Biomedical Engineering, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey
| | - Fatma Dogan Guzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, 06010 Ankara, Turkey;
| | - Ali Akpek
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Gözde Yesiltas
- Department of Bioengineering, Gebze Technical University, 41400 Gebze, Turkey; (A.A.); (G.Y.)
| | - Tuba Canak-Ipek
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany;
| |
Collapse
|
16
|
Kapoor S, Subba P, Shenoy P S, Bose B. Sca1 + Progenitor Cells (Ex vivo) Exhibits Differential Proteomic Signatures From the Culture Adapted Sca1 + Cells (In vitro), Both Isolated From Murine Skeletal Muscle Tissue. Stem Cell Rev Rep 2021; 17:1754-1767. [PMID: 33742350 DOI: 10.1007/s12015-021-10134-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 10/21/2022]
Abstract
Stem cell antigen-1 (Sca-1) is a glycosyl-phosphatidylinositol-anchored membrane protein that is expressed in a sub-population of muscle stem and progenitor cell types. Reportedly, Sca-1 regulates the myogenic property of myoblasts and Sca-1-/- mice exhibited defective muscle regeneration. Although the role of Sca-1 in muscle development and maintenance is well-acknowledged, molecular composition of muscle derived Sca-1+ cells is not characterized. Here, we applied a high-resolution mass spectrometry-based workflow to characterize the proteomic landscape of mouse hindlimb skeletal muscle derived Sca-1+ cells. Furthermore, we characterized the impact of the cellular microenvironments on the proteomes of Sca-1+ cells. The proteome component of freshly isolated Sca-1+ cells (ex vivo) was compared with that of Sca-1+ cells expanded in cell culture (in vitro). The analysis revealed significant differences in the protein abundances in the two conditions reflective of their functional variations. The identified proteins were enriched in various biological pathways. Notably, we identified proteins related to myotube differentiation, myotube cell development and myoblast fusion. We also identified a panel of cell surface marker proteins that can be leveraged in future to enrich Sca-1+ cells using combinatorial strategies. Comparative analysis implicated the activation of various pathways leading to increased protein synthesis under in vitro condition. We report here the most comprehensive proteome map of Sca-1+ cells that provides insights into the molecular networks operative in Sca-1+ cells. Importantly, through our work we generated the proteomic blueprint of protein abundances significantly altered in Sca-1+ cells under ex vivo and in vitro conditions. The curated data can also be visualized at https://yenepoya.res.in/database/Sca-1-Proteomics .
Collapse
Affiliation(s)
- Saketh Kapoor
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Pratigya Subba
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka, 575018, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
17
|
García-Prat L, Perdiguero E, Alonso-Martín S, Dell'Orso S, Ravichandran S, Brooks SR, Juan AH, Campanario S, Jiang K, Hong X, Ortet L, Ruiz-Bonilla V, Flández M, Moiseeva V, Rebollo E, Jardí M, Sun HW, Musarò A, Sandri M, Del Sol A, Sartorelli V, Muñoz-Cánoves P. FoxO maintains a genuine muscle stem-cell quiescent state until geriatric age. Nat Cell Biol 2020; 22:1307-1318. [PMID: 33106654 DOI: 10.1038/s41556-020-00593-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Tissue regeneration declines with ageing but little is known about whether this arises from changes in stem-cell heterogeneity. Here, in homeostatic skeletal muscle, we identify two quiescent stem-cell states distinguished by relative CD34 expression: CD34High, with stemness properties (genuine state), and CD34Low, committed to myogenic differentiation (primed state). The genuine-quiescent state is unexpectedly preserved into later life, succumbing only in extreme old age due to the acquisition of primed-state traits. Niche-derived IGF1-dependent Akt activation debilitates the genuine stem-cell state by imposing primed-state features via FoxO inhibition. Interventions to neutralize Akt and promote FoxO activity drive a primed-to-genuine state conversion, whereas FoxO inactivation deteriorates the genuine state at a young age, causing regenerative failure of muscle, as occurs in geriatric mice. These findings reveal transcriptional determinants of stem-cell heterogeneity that resist ageing more than previously anticipated and are only lost in extreme old age, with implications for the repair of geriatric muscle.
Collapse
Affiliation(s)
- Laura García-Prat
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eusebio Perdiguero
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Sonia Alonso-Martín
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Neurosciences Area, Biodonostia Health Research Institute, Donostia-San Sebastián, San Sebastián, Spain
| | - Stefania Dell'Orso
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Srikanth Ravichandran
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Stephen R Brooks
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA
| | - Silvia Campanario
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Kan Jiang
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Xiaotong Hong
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain
| | - Laura Ortet
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Vanessa Ruiz-Bonilla
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Marta Flández
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.,Grupo de Investigación en Oncología Clínico Traslacional, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Victoria Moiseeva
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Mercè Jardí
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH Bethesda, Bethesda, MD, USA
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Marco Sandri
- Department of Biomedical Science, University of Padova, Padova, Italy
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.,CIC bioGUNE, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH Bethesda, Bethesda, MD, USA.
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain. .,Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
18
|
Huang S, Jin M, Su N, Chen L. New insights on the reparative cells in bone regeneration and repair. Biol Rev Camb Philos Soc 2020; 96:357-375. [PMID: 33051970 DOI: 10.1111/brv.12659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique characteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years, there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have greatly hampered our understanding of these important cell populations and their potential applications in the treatment of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and pericytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in this field.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| |
Collapse
|
19
|
May V, Arnold AA, Pagad S, Somagutta MR, Sridharan S, Nanthakumaran S, Malik BH. Duchenne's Muscular Dystrophy: The Role of Induced Pluripotent Stem Cells and Genomic Editing on Muscle Regeneration. Cureus 2020; 12:e10600. [PMID: 33123420 PMCID: PMC7584317 DOI: 10.7759/cureus.10600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
There are two types of well-known muscular dystrophies: Duchenne's muscular dystrophy (DMD) and Becker's muscular dystrophy. This article focuses on the X-linked recessive disorder of Duchenne's muscular dystrophy, which primarily affects children at age four, with a shortened life span of up to 40 years. A defective dystrophin protein lacking the gene dystrophin is the primary cause of the disease pathophysiology. This defect causes cardiac and skeletal muscle down-regulation of dystrophin, leading to weak and fibrotic muscles. The disease is currently untreatable, so most kids die due to cardiac failure in their late 30's. This review presents current treatment options, based on previous studies conducted over the last five years. We used the PubMed database to analyze and review the most important investigations. We also included an analysis of induced pluripotent stem cell therapy vs. genetic therapy using the mdx mouse model. We have discovered promising results on mdx mouse models to date and excited about the potential for where further clinical human trials can go.
Collapse
Affiliation(s)
- Vanessa May
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ashley A Arnold
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sukrut Pagad
- Department of Internal Medicine, Larkin Community Hospital, Hialeah, USA
| | - Manoj R Somagutta
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saijanakan Sridharan
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saruja Nanthakumaran
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bilal Haider Malik
- Department of Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
20
|
Silva CGD, Barretto LSDS, Lo Turco EG, Santos ADL, Lessio C, Martins Júnior HA, Almeida FGD. Lipidomics of mesenchymal stem cell differentiation. Chem Phys Lipids 2020; 232:104964. [PMID: 32882223 DOI: 10.1016/j.chemphyslip.2020.104964] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/31/2020] [Accepted: 08/23/2020] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSCs), such as adipose-derived stem cells (ADSCs) and skeletal muscle-derived stem cells (MDSCs), are potential sources for cell-based therapeutic strategies. However, there is little knowledge about the lipid composition of these stem cells and the mechanisms of their differentiation. Lipids have important biological and physiological functions that are critical for understanding the regulation and control of stem cell fate. This study sought to analyze the lipidome of rabbit ADSCs and MDSCs and their adipogenic and osteogenic differentiation. The MSCs were isolated and were characterized by flow cytometry. Lipids were extracted from both MSCs and differentiated cells, and the lipids were subsequently analyzed with a hybrid triple quadrupole time-of-flight mass spectrometer. The results showed a total of 1687 lipid species. MSCs exhibited different lipid profiles as well as changes in lipid composition after differentiation. Furthermore, the expression levels of N-acyl-phosphatidylethanolamine (NAPE) 54:7+NH4 (-FA 17:0(NH4)) and phosphatidylcholine (PC) 42:6+Na were higher in the adipogenic lineages in of both MSC types, and NAPE 58:2+NH4 (-FA 17:0 (NH4)) and NAPE 56:2+NH4 (-FA 17:0 (NH4)) had higher levels in the osteogenic lineages, suggesting lipid similarities in cells differentiated from different stem cell sources.
Collapse
Affiliation(s)
- Camila Gonzaga da Silva
- Department of Surgery, Division of Urology, Federal University of São Paulo, Rua Embaú 231- Vila Clementino, São Paulo, SP, 04039-060, Brazil
| | - Letícia Siqueira de Sá Barretto
- Department of Surgery, Division of Urology, Federal University of São Paulo, Rua Embaú 231- Vila Clementino, São Paulo, SP, 04039-060, Brazil.
| | - Edson Guimarães Lo Turco
- Department of Surgery, Division of Urology, Federal University of São Paulo, Rua Embaú 231- Vila Clementino, São Paulo, SP, 04039-060, Brazil
| | - Alex de Lima Santos
- Department of Surgery, Division of Urology, Federal University of São Paulo, Rua Embaú 231- Vila Clementino, São Paulo, SP, 04039-060, Brazil
| | - Camila Lessio
- Department of Surgery, Division of Urology, Federal University of São Paulo, Rua Embaú 231- Vila Clementino, São Paulo, SP, 04039-060, Brazil
| | | | - Fernando Gonçalves de Almeida
- Department of Surgery, Division of Urology, Federal University of São Paulo, Rua Embaú 231- Vila Clementino, São Paulo, SP, 04039-060, Brazil
| |
Collapse
|
21
|
Tamaki T. Biomedical applications of muscle-derived stem cells: from bench to bedside. Expert Opin Biol Ther 2020; 20:1361-1371. [PMID: 32643444 DOI: 10.1080/14712598.2020.1793953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Skeletal muscle-derived stem cells (Sk-MDSCs) are considered promising sources of adult stem cell therapy. Skeletal muscle comprises approximately 40-50% of the total body mass with marked potential for postnatal adaptive response, such as muscle hypertrophy, hyperplasia, atrophy, and regenerative capacity. This strongly suggests that skeletal muscle contains various stem/progenitor cells related to muscle-nerve-vascular tissues, which would support the above postnatal events even in adulthood. AREA COVERED The focus of this review is the therapeutic potential of the Sk-MDSCs as an adult stem cell autograft. For this purpose, the validity of cell isolation and purification, tissue reconstitution capacity in vivo after transplantation, comparison of the results of basic mouse and preclinical human studies, potential problematic and beneficial aspects, and effective usage have been discussed following the history of clinical applications. EXPERT OPINION Although the clinical application of Sk-MDSCs began as a therapy for the systemic disease of Duchenne muscular dystrophy, here, through the unique local injection method, therapy for severely damaged peripheral nerves, particularly the long-gap nerve transection, has been introduced. The beneficial aspects of the use of Sk-MDSCs as the source of local tissue transplantation therapy have also been discussed.
Collapse
Affiliation(s)
- Tetsuro Tamaki
- Muscle Physiology and Cell Biology Unit, Department of Physiology, Tokai University School of Medicine , Isehara, Kanagawa ,Japan
| |
Collapse
|
22
|
Nakayama N, Pothiawala A, Lee JY, Matthias N, Umeda K, Ang BK, Huard J, Huang Y, Sun D. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020; 77:2543-2563. [PMID: 31915836 PMCID: PMC11104892 DOI: 10.1007/s00018-019-03445-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The cartilage of joints, such as meniscus and articular cartilage, is normally long lasting (i.e., permanent). However, once damaged, especially in large animals and humans, joint cartilage is not spontaneously repaired. Compensating the lack of repair activity by supplying cartilage-(re)forming cells, such as chondrocytes or mesenchymal stromal cells, or by transplanting a piece of normal cartilage, has been the basis of therapy for biological restoration of damaged joint cartilage. Unfortunately, current biological therapies face problems on a number of fronts. The joint cartilage is generated de novo from a specialized cell type, termed a 'joint progenitor' or 'interzone cell' during embryogenesis. Therefore, embryonic chondroprogenitors that mimic the property of joint progenitors might be the best type of cell for regenerating joint cartilage in the adult. Pluripotent stem cells (PSCs) are expected to differentiate in culture into any somatic cell type through processes that mimic embryogenesis, making human (h)PSCs a promising source of embryonic chondroprogenitors. The major research goals toward the clinical application of PSCs in joint cartilage regeneration are to (1) efficiently generate lineage-specific chondroprogenitors from hPSCs, (2) expand the chondroprogenitors to the number needed for therapy without loss of their chondrogenic activity, and (3) direct the in vivo or in vitro differentiation of the chondroprogenitors to articular or meniscal (i.e., permanent) chondrocytes rather than growth plate (i.e., transient) chondrocytes. This review is aimed at providing the current state of research toward meeting these goals. We also include our recent achievement of successful generation of "permanent-like" cartilage from long-term expandable, hPSC-derived ectomesenchymal chondroprogenitors.
Collapse
Affiliation(s)
- Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA.
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - Katsutsugu Umeda
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Department of Pediatrics, Kyoto University School of Medicine, Kyoto, Japan
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yun Huang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Deqiang Sun
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
23
|
IL-4 and SDF-1 Increase Adipose Tissue-Derived Stromal Cell Ability to Improve Rat Skeletal Muscle Regeneration. Int J Mol Sci 2020; 21:ijms21093302. [PMID: 32392778 PMCID: PMC7246596 DOI: 10.3390/ijms21093302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle regeneration depends on the satellite cells, which, in response to injury, activate, proliferate, and reconstruct damaged tissue. However, under certain conditions, such as large injuries or myopathies, these cells might not sufficiently support repair. Thus, other cell populations, among them adipose tissue-derived stromal cells (ADSCs), are tested as a tool to improve regeneration. Importantly, the pro-regenerative action of such cells could be improved by various factors. In the current study, we tested whether IL-4 and SDF-1 could improve the ability of ADSCs to support the regeneration of rat skeletal muscles. We compared their effect at properly regenerating fast-twitch EDL and poorly regenerating slow-twitch soleus. To this end, ADSCs subjected to IL-4 and SDF-1 were analyzed in vitro and also in vivo after their transplantation into injured muscles. We tested their proliferation rate, migration, expression of stem cell markers and myogenic factors, their ability to fuse with myoblasts, as well as their impact on the mass, structure and function of regenerating muscles. As a result, we showed that cytokine-pretreated ADSCs had a beneficial effect in the regeneration process. Their presence resulted in improved muscle structure and function, as well as decreased fibrosis development and a modulated immune response.
Collapse
|
24
|
Dong Y, Li Y, Zhang C, Chen H, Liu L, Chen S. Effects of SW033291 on the myogenesis of muscle-derived stem cells and muscle regeneration. Stem Cell Res Ther 2020; 11:76. [PMID: 32085799 PMCID: PMC7035785 DOI: 10.1186/s13287-020-1574-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background The unmet medical needs in repairing large muscle defects promote the development of tissue regeneration strategy. The use of bioactive molecules in combination with biomaterial scaffold has become an area of great interest. SW033291, a small-molecule inhibitor targeting 15-hydroxyprostaglandin dehydrogenase (15-PDGH) and subsequently elevating the production of prostaglandin E2 (PGE2), has been proved to accelerate the recovery and potentiate the regeneration of multiple tissues including the bone, liver, and colon. The limited understanding of the potential therapeutic effects on myogenesis motivated us to investigate the role of SW033291 in regulating muscle-derived stem cell (MDSC) myogenic differentiation and MDSC-mediated muscle regeneration. Methods The characteristics of rat MDSCs, including cell-specific markers and myogenic differentiation potential, were determined. MDSCs were incubated with SW033291 to evaluate PGE2 production and cytotoxicity. The effects of SW033291 on MDSC myogenic differentiation were assessed by quantitative real-time polymerase chain reaction (qPCR), western blot, and immunocytochemistry. The fibrin gel containing MDSCs and SW033291 was used for muscle regeneration in a tibialis anterior muscle defect model. Results Our data demonstrated that MDSCs were well-tolerated to SW033291 and treatment with SW033291 significantly promoted the production of PGE2 by MDSCs. In vitro analysis showed that SW033291 enhanced the myogenic differentiation and myotube formation by upregulating a series of myogenic markers. Additionally, the activation of PI3K/Akt pathway was involved in the mechanism underlying these promotive effects. Then, in situ casting of fibrin gel containing MDSCs and SW033291 was used to repair the tibialis anterior muscle defect; the addition of SW033291 significantly promoted myofiber formation within the defect region with mild immune response, less fibrosis, and sufficient vascularization. Conclusion SW033291 acted as a positive regulator of MDSC myogenic differentiation, and incorporating the compound with MDSCs in fibrin gel could serve as an effective method to repair large skeletal muscle defects.
Collapse
Affiliation(s)
- Yuanqiang Dong
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yuan Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Chuan Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Haibin Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Lijia Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Simeng Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
25
|
Regenerative medicine and injection therapies in stress urinary incontinence. Nat Rev Urol 2020; 17:151-161. [PMID: 31974507 DOI: 10.1038/s41585-019-0273-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2019] [Indexed: 02/06/2023]
Abstract
Stress urinary incontinence (SUI) is a common and bothersome condition. Anti-incontinence surgery has high cure rates, but concerns about mesh tapes have resulted in the resurgence of surgical procedures that involve increased abdominopelvic dissection and morbidity. Injection therapy with urethral bulking agents or stem cell formulations have been developed as minimally invasive alternatives. Many synthetic and biological bulking agents have been trialled, but several have been discontinued owing to safety concerns. The use of Macroplastique and Contigen has the largest evidence base, but, overall, success rates seem to be similar between the various agents and positive outcomes are poorly sustained for more than 6 months. Furthermore, subjective cure rates, although initially high, also deteriorate over time. The available data consistently demonstrate manifestly poorer outcomes for injection therapies than for surgery. Stem cell treatments are thought to functionally regenerate the urethral sphincter in patients with suspected intrinsic sphincter deficiency. Autologous adipose and muscle-derived stem cells seem to be the intuitive cell source, as they are comparatively abundant, can be harvested and cause minimal donor site morbidity. To date, only a few small clinical studies have been reported and most data are derived from animal models. The success rates of stem cell injection therapies seem to be comparable with those of bulking agents.
Collapse
|
26
|
Whiting D, Hamdoon M, Sriprasad S. Stem cell therapy for stress urinary incontinence. JOURNAL OF CLINICAL UROLOGY 2020. [DOI: 10.1177/2051415819841957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Stress urinary incontinence is the involuntary loss of urine on effort or physical exertion. It is a highly prevalent condition affecting both men and women. Treatment is performed in a step-wise approach involving conservative measures, such as weight loss and pelvic floor exercises, medical treatment with duloxetine and a variety of surgical treatment options. However, recent restrictions in the use of synthetic mesh and tape have limited the surgical treatment options, leading to the need for new and novel treatment for stress urinary incontinence. Stem cell therapy is a developing medical field and offers the potential to restore normal physiological function of the urethral sphincter. The effectiveness of stem cell therapy in stress urinary incontinence has been demonstrated in pre-clinical studies, leading to its evaluation in several clinical studies. This review assesses the current evidence for the safety and efficacy of stem cell treatment for patients with stress urinary incontinence who have failed conservative and/or medical management and have not undergone previous surgical treatment for stress urinary incontinence.Evidence Level: Not applicable
Collapse
Affiliation(s)
- D Whiting
- Department of Urology, Darent Valley Hospital, United Kingdom
| | - M Hamdoon
- Department of Urology, Darent Valley Hospital, United Kingdom
| | - S Sriprasad
- Department of Urology, Darent Valley Hospital, United Kingdom
| |
Collapse
|
27
|
Kapoor S, Shenoy SP, Bose B. CD34 cells in somatic, regenerative and cancer stem cells: Developmental biology, cell therapy, and omics big data perspective. J Cell Biochem 2019; 121:3058-3069. [PMID: 31886574 DOI: 10.1002/jcb.29571] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
The transmembrane phosphoglycoprotein protein CD34 has conventionally been regarded as a marker for hematopoietic progenitors. Its expression on these cells has been leveraged for cell therapy applications in various hematological disorders. More recently, the expression of CD34 has also been reported on cells of nonhematopoietic origin. The list includes somatic cells such as endothelial cells, fibrocytes and interstitial cells and regenerative stem cells such as corneal keratocytes, muscle satellite cells, and muscle-derived stem cells. Furthermore, its expression on some cancer stem cells (CSCs) has also been reported. Till date, the functional roles of this molecule have been implicated in a multitude of cellular processes including cell adhesion, signal transduction, and maintenance of progenitor phenotype. However, the complete understanding about this molecule including its developmental origins, its embryonic connection, and associated functions is far from complete. Here, we review our present understanding of the structure and putative functions of the CD34 molecule based upon our literature survey. We also probed various biological databases to retrieve data related to the expression and associated molecular functions of CD34. Such information, upon synthesis, is hence likely to provide the suitability of such cells for cell therapy. Moreover, we have also covered the existing cell therapy and speculated cell therapy applications of CD34+ cells isolated from various lineages. We have also attempted here to speculate the role(s) of CD34 on CSCs. Finally, we discuss number of large-scale proteomics and transcriptomics studies that have been performed using CD34+ cells.
Collapse
Affiliation(s)
- Saketh Kapoor
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sudheer P Shenoy
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| |
Collapse
|
28
|
Xu Z, Chen Z, Feng W, Huang M, Yang X, Qi Z. Grafted muscle-derived stem cells promote the therapeutic efficiency of epimysium conduits in mice with peripheral nerve gap injury. Artif Organs 2019; 44:E214-E225. [PMID: 31792982 DOI: 10.1111/aor.13614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/25/2019] [Accepted: 11/29/2019] [Indexed: 12/24/2022]
Abstract
Our research aimed to build allogeneic artificial conduits with epimysium and muscle-derived stem cells (MDSCs) from the skeletal muscle of mice. We applied the conduit to repair peripheral nerve defects and estimated the effectiveness of the repair process. In the research, we prepared epimysium conduits with lumens to bridge repair a 5-mm-long sciatic nerve defect from C57 wild-type mice and then transplanted green fluorescent protein (GFP)-MDSCs and Matrigel suspensions into the conduit. Histological and functional assessments were performed 4 and 8 weeks after surgery. The tissue-engineered conduit from muscle effectively repaired the nerve defect, while the group with GFP-MDSCs showed improved histological examinations and functional assessments, and the newborn nerves highly expressed GFP. As the results suggested, autologous epimysium conduits represent a reliable method to repair peripheral nerve defects, and the addition of MDSCs promote the effectiveness of differentiating into multiple lineages. Our research simultaneously demonstrated the myogenic, neurogenic, and angiogenic potential of MDSCs in vivo for the first time.
Collapse
Affiliation(s)
- Zhuqiu Xu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zixiang Chen
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weifeng Feng
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minlu Huang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaonan Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zuoliang Qi
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
29
|
Bayrak Ö, Dmochowski RR. Underactive bladder: A review of the current treatment concepts. Turk J Urol 2019; 45:401-409. [PMID: 30817271 PMCID: PMC6788564 DOI: 10.5152/tud.2019.37659] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 01/31/2023]
Abstract
According to the International Continence Society standardization reports, underactive bladder (UAB) is a decrease in detrusor contraction and/or shortening of the contraction time, resulting in an incomplete and/or prolongation of the bladder emptying within the normal time frame. It has been indicated that idiopathic, neurogenic, myogenic, and iatrogenic factors play a role in the etiology. To make a diagnosis, it is absolutely necessary to perform a pressure-flow study. Treatment alternatives are generally based on the evacuation of the lower urinary tract, independent of the etiology. UAB treatments are listed under the headings of conservative methods and clean intermittent catheterization, pharmacotherapy (alpha-blockers, cholinesterase inhibitors, muscarinic agonists, prostaglandin E2, and acotiamide), surgical treatments (sacral nerve stimulation-electrical stimulation, injections into the external sphincter, surgeries to be performed for bladder outlet obstruction, reduction cystoplasty, and latissimus dorsi detrusor myoplasty), and stem cell and gene therapies. It is still controversial whether satisfactory success is achieved in the treatment of patients with UAB. Owing to the better understanding of the pathophysiology, future developments in the pharmaceutical industry, gene therapy, and biomedical applications are expected to close the gap in the treatment.
Collapse
Affiliation(s)
- Ömer Bayrak
- Department of Urology, Gaziantep University School of Medicine, Gaziantep, Turkey
| | | |
Collapse
|
30
|
Peng D, Yuan H, Liu T, Wang T, Reed-Maldonado AB, Kang N, Banie L, Wang G, Tang Y, He L, Lin G, Lue TF. Smooth Muscle Differentiation of Penile Stem/Progenitor Cells Induced by Microenergy Acoustic Pulses In Vitro. J Sex Med 2019; 16:1874-1884. [PMID: 31585805 DOI: 10.1016/j.jsxm.2019.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Modulating tissue-resident stem and progenitor cells with a non-invasive, mechanobiological intervention is an optimal approach for tissue regeneration. Stem cell antigen-1 (Sca-1) has been identified as a stem cell marker within many organs but never within the penis. AIM To localize and isolate penile stem/progenitor cells (PSPCs) and to evaluate cellular differentiation after exposure to induction medium and microenergy acoustic pulse (MAP) therapy. METHODS Six male Sprague-Dawley rats were used to isolate PSPCs. Isolation was followed by stem cell characterization and differentiation assays. The PSPCs were then treated with MAP (0.033 mJ/mm2, 1 Hz) at various dosages (25, 50, 100, and 200 pulses) and for different durations (1, 2, 4, 6, or 8 hours) in vitro. MAIN OUTCOME MEASURE The PSPCs (Sca-1-positive cells) were isolated using the magnetic-activated cell sorting system. PSPC cellular differentiation was assessed after induction with induction medium and with MAP in vitro. Wnt/β-catenin signaling was also assayed. RESULTS The PSPCs were successfully localized within the penile subtunic and perisinusoidal spaces, and they were successfully isolated using magnetic-activated cell sorting. The stemness of the cells was confirmed by stem cell marker characterization and by multiple differentiation into smooth muscle cells, endothelial cells, adipocytes, and neurons. MAP-induced PSPCs differentiated into smooth muscle cells by activating the Wnt/β-catenin signaling pathway in a time- and dosage-dependent manner. CLINICAL IMPLICATIONS By modulating resident PSPCs, MAP may have utility in the treatment of erectile dysfunction (ED). STRENGTHS & LIMITATIONS This study provides solid evidence in support of microenergy therapies, including both MAP and low-intensity extracorporeal shock wave therapy, for the treatment of ED. Additional studies are needed and should include additional stem cells markers. Furthermore, studies exploring the underling mechanisms for PSPC activation and differentiation are required. CONCLUSION PSPCs were successfully identified, localized, and isolated. Additionally, MAP provoked PSPCs to differentiate into smooth muscle cells via the Wnt/β-catenin signaling pathway. As such, MAP provides a novel method for activating endogenous tissue-resident stem/progenitor cells and might facilitate stem cell regenerative therapy targeting ED. Peng D, Yuan H, Liu T, et al. Smooth Muscle Differentiation of Penile Stem/Progenitor Cells Induced by Microenergy Acoustic Pulses In Vitro. J Sex Med 2019; 16:1874-1884.
Collapse
Affiliation(s)
- Dongyi Peng
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA; Department of Urology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Huixing Yuan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tianshu Liu
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tianyu Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Amanda B Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Ning Kang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Yuxin Tang
- Department of Urology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Leye He
- Department of Urology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
31
|
Grabowska I, Zimowska M, Maciejewska K, Jablonska Z, Bazga A, Ozieblo M, Streminska W, Bem J, Brzoska E, Ciemerych MA. Adipose Tissue-Derived Stromal Cells in Matrigel Impacts the Regeneration of Severely Damaged Skeletal Muscles. Int J Mol Sci 2019; 20:E3313. [PMID: 31284492 PMCID: PMC6651806 DOI: 10.3390/ijms20133313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 02/07/2023] Open
Abstract
In case of large injuries of skeletal muscles the pool of endogenous stem cells, i.e., satellite cells, might be not sufficient to secure proper regeneration. Such failure in reconstruction is often associated with loss of muscle mass and excessive formation of connective tissue. Therapies aiming to improve skeletal muscle regeneration and prevent fibrosis may rely on the transplantation of different types of stem cell. Among such cells are adipose tissue-derived stromal cells (ADSCs) which are relatively easy to isolate, culture, and manipulate. Our study aimed to verify applicability of ADSCs in the therapies of severely injured skeletal muscles. We tested whether 3D structures obtained from Matrigel populated with ADSCs and transplanted to regenerating mouse gastrocnemius muscles could improve the regeneration. In addition, ADSCs used in this study were pretreated with myoblasts-conditioned medium or anti-TGFβ antibody, i.e., the factors modifying their ability to proliferate, migrate, or differentiate. Analyses performed one week after injury allowed us to show the impact of 3D cultured control and pretreated ADSCs at muscle mass and structure, as well as fibrosis development immune response of the injured muscle.
Collapse
Affiliation(s)
- Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Malgorzata Zimowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Karolina Maciejewska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Zuzanna Jablonska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Anna Bazga
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Michal Ozieblo
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Wladyslawa Streminska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Joanna Bem
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland.
| |
Collapse
|
32
|
|
33
|
Scibetta AC, Morris ER, Liebowitz AB, Gao X, Lu A, Philippon MJ, Huard J. Characterization of the chondrogenic and osteogenic potential of male and female human muscle-derived stem cells: Implication for stem cell therapy. J Orthop Res 2019; 37:1339-1349. [PMID: 30667562 DOI: 10.1002/jor.24231] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/02/2019] [Indexed: 02/04/2023]
Abstract
People of all backgrounds are susceptible to bone and cartilage damage, and these injuries can be debilitating. Current treatments for bone and cartilage injuries are less than optimal, and we are interested in developing new approaches to treat these diseases, specifically using human muscle-derived stem cells (hMDSCs). Our lab previously demonstrated that sex differences exist between male and female murine MDSCs; thus, this paper sought to investigate whether sex differences also exist in hMDSCs. In the present study, we characterized the chondrogenic and osteogenic sex differences of hMDSCs in vitro and in vivo. We performed in vitro osteogenic and chondrogenic differentiation using hMDSC pellet cultures. As demonstrated by microCT, histology, and immunohistochemistry, male hMDSCs were more chondrogenic and osteogenic than their female counterparts in vitro. No differences were observed based on the sex of hMDSCs in osteogenic and chondrogenic gene expression and cell surface markers. For our in vivo study, we transduced hMDSCs with lenti-BMP2/GFP and transplanted these cells into critical-sized calvarial defects in mice. MicroCT results revealed that male hMDSCs regenerated more bone at 2 weeks and demonstrated higher bone density at 4 and 6 weeks than female hMDSCs. Histology demonstrated that both male and female hMDSCs regenerated functional bone. Clinical relevance: These studies reinforce that stem cells isolated from male and female patients differ in function, and we should disclose the sex of cells used in future studies. Considering sex differences of hMDSCs may help to improve cell-based therapies for autologous cell treatment of bone and cartilage damage. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1339-1349, 2019.
Collapse
Affiliation(s)
| | | | | | - Xueqin Gao
- Steadman Philippon Research Institute, Vail, Colorado.,Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Aiping Lu
- Steadman Philippon Research Institute, Vail, Colorado.,Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | | | - Johnny Huard
- Steadman Philippon Research Institute, Vail, Colorado.,Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
34
|
Liu G, Liao C, Chen X, Xu Y, Tan J, Han F, Ye X. Identification and Characterization of Skeletal Muscle Stem Cells from Human Orbicularis Oculi Muscle. Tissue Eng Part C Methods 2019; 24:486-493. [PMID: 29993336 DOI: 10.1089/ten.tec.2018.0048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Skeletal muscle stem cell (SMSC) transplantation has shown great therapeutical potential in repairing muscle loss and dysfunction, but the muscle acquisition is usually a traumatic procedure causing pain and morbidity to the donor. In this study, we investigated the feasibility of isolating SMSCs from human orbicularis oculi muscle (OOM), which is routinely removed and discarded during ophthalmic cosmetic surgeries. OOM fragments were harvested from 18 female healthy donors undergoing upper eyelid plasties. Plastic-adherent cells were isolated from the muscles using a two-step plating method combined with collagenase digestion. A total of 15 cell cultures were successfully established from the muscle samples. These adherent cells were positive for the specific markers of SMSCs and could be directed toward the osteogenic, adipogenic, chondrogenic, and myogenic phenotypes in the presence of lineage-specific inductive media. Moreover, after cultured in the myogenic inductive medium for 3 weeks, the muscle cells were injected into the tibialis anterior muscles of nude mice and the cell fate was detected using a DiI-labeling technique. In vivo myogenesis was evidenced by the expression of DiI fluorescence after cell transplantation. The donor cells could be found in the satellite cell position and incorporated into the host myofibers. Our results demonstrated that human OOM represents a novel source of myogenic precursors with stem cell-like properties, which may provide a foundation for the SMSC-based therapeutics of skeletal muscle diseases.
Collapse
Affiliation(s)
- Guangpeng Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Caihe Liao
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Xi Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Yipin Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Jian Tan
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Fang Han
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| | - Xinhai Ye
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai, China
| |
Collapse
|
35
|
Muscle-Derived Stem Cell-Enriched Scaffolds Are Capable of Enhanced Healing of a Murine Volumetric Muscle Loss Defect. Plast Reconstr Surg 2019; 143:329e-339e. [PMID: 30531618 DOI: 10.1097/prs.0000000000005273] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Volumetric muscle loss secondary to traumatic or surgical causes can lead to functional and aesthetic impairments. The authors hypothesize that an implantable muscle-derived stem cell-enriched collagen scaffold could significantly augment muscle regeneration in a murine model of volumetric muscle loss. METHODS Murine muscle-derived stem cells were isolated using a modified preplating technique and seeded onto type 1 collagen scaffolds to create the muscle-derived stem cell-enriched collagen scaffolds. Murine rectus femoris defects of 5 mm were created and randomized to one of three conditions (n = 6 per group): untreated controls, collagen scaffold only, and muscle-derived stem cell-enriched collagen scaffolds. In vivo muscle healing was quantified using micro-computed tomography. Muscle explants were analyzed using standard histology and whole-mount immunofluorescence at 8 weeks. RESULTS In vivo experiments demonstrated significantly greater quadriceps cross-sectional area in the muscle-derived stem cell-enriched collagen scaffold group compared with controls on micro-computed tomography (0.74 ± 0.21 versus 0.55 ± 0.06 versus 0.49 ± 0.04 ratio of experimental to naive quadriceps cross-sectional area; p < 0.05). Muscle explants of the muscle-derived stem cell-enriched collagen scaffold group demonstrated significantly higher cellular density compared with controls (1185 ± 360 versus 359 ± 62 versus 197 ± 68 nuclei/high-power field; p < 0.01). Immunofluorescence for laminin and myosin heavy chain confirmed formation of organized muscle fibers within the defect of the muscle-derived stem cell-enriched collagen scaffold group only. However, appreciable confocal colocalization of myosin heavy chain with green fluorescent protein expression was low. CONCLUSIONS The results of this study indicate that muscle-derived stem cell-enriched scaffolds significantly improved skeletal muscle regeneration in a murine muscle defect model. Based on the low fluorescent colocalization, host progenitor cells appear to contribute significantly to intradefect myogenesis, suggesting that deployment of a viable muscle-derived stem cell-enriched scaffold stimulates a regenerative mitogen response in native tissues.
Collapse
|
36
|
Tang X, Daneshmandi L, Awale G, Nair LS, Laurencin CT. Skeletal Muscle Regenerative Engineering. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019; 5:233-251. [PMID: 33778155 DOI: 10.1007/s40883-019-00102-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Skeletal muscles have the intrinsic ability to regenerate after minor injury, but under certain circumstances such as severe trauma from accidents, chronic diseases or battlefield injuries the regeneration process is limited. Skeletal muscle regenerative engineering has emerged as a promising approach to address this clinical issue. The regenerative engineering approach involves the convergence of advanced materials science, stem cell science, physical forces, insights from developmental biology, and clinical translation. This article reviews recent studies showing the potential of the convergences of technologies involving biomaterials, stem cells and bioactive factors in concert with clinical translation, in promoting skeletal muscle regeneration. Several types of biomaterials such as electrospun nanofibers, hydrogels, patterned scaffolds, decellularized tissues, and conductive matrices are being investigated. Detailed discussions are given on how these biomaterials can interact with cells and modulate their behavior through physical, chemical and mechanical cues. In addition, the application of physical forces such as mechanical and electrical stimulation are reviewed as strategies that can further enhance muscle contractility and functionality. The review also discusses established animal models to evaluate regeneration in two clinically relevant muscle injuries; volumetric muscle loss (VML) and muscle atrophy upon rotator cuff injury. Regenerative engineering approaches using advanced biomaterials, cells, and physical forces, developmental cues along with insights from immunology, genetics and other aspects of clinical translation hold significant potential to develop promising strategies to support skeletal muscle regeneration.
Collapse
Affiliation(s)
- Xiaoyan Tang
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Guleid Awale
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT 06030, USA.,Department of Orthopaedic Surgery, UConn Health, Farmington, CT 06030, USA.,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA.,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA.,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
37
|
Biz C, Crimi A, Fantoni I, Pozzuoli A, Ruggieri P. Muscle stem cells: what's new in orthopedics? ACTA BIO-MEDICA : ATENEI PARMENSIS 2019; 90:8-13. [PMID: 30714993 PMCID: PMC6503412 DOI: 10.23750/abm.v90i1-s.8078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIM OF THE WORK Adult stem cells were studied as a source of potentially useful development for tissue engineering and repair techniques. The aim of this review is to clarify the actual and possible uses of muscle stem cells in orthopedics. METHODS A selection of studies was made to obtain a homogeneous and up to date overview on the muscle stem cells applications. RESULTS In recent years muscle was studied as a good source of adult stem cells that can differentiate into different cell lineages. Muscle stem cells are a heterogeneous population of cells, which demonstrated in vitro a great potential for the regeneration and repair of muscle, bone and cartilage tissue. Among muscle stem cells, satellite stem cells are the most known progenitor cells: they can differentiate in osteoblasts, adipocytes, chondrocytes and myocytes. CONCLUSIONS Although muscle stem cells are a promising field of research, more pre-clinical studies in animal models are still needed to determine the safety and efficiency of the transplant procedures in humans.
Collapse
Affiliation(s)
- Carlo Biz
- Orthopaedic Clinic, Department of Surgery, Oncology and Gastroenterology DiSCOG, University of Padua, Padova, Italy.
| | | | | | | | | |
Collapse
|
38
|
Tseng C, Sinha K, Pan H, Cui Y, Guo P, Lin CY, Yang F, Deng Z, Eltzschig HK, Lu A, Huard J. Markers of Accelerated Skeletal Muscle Regenerative Response in Murphy Roths Large Mice: Characteristics of Muscle Progenitor Cells and Circulating Factors. Stem Cells 2019; 37:357-367. [PMID: 30537304 DOI: 10.1002/stem.2957] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/06/2018] [Accepted: 11/14/2018] [Indexed: 12/19/2022]
Abstract
The "super-healing" Murphy Roths Large (MRL/MpJ) mouse possesses a superior regenerative capacity for repair of many tissues, which makes it an excellent animal model for studying molecular and cellular mechanisms during tissue regeneration. As the role of muscle progenitor cells (MPCs) in muscle-healing capacity of MRL/MpJ mice has not been previously studied, we investigated the muscle regenerative capacity of MRL/MpJ mice following muscle injury, and the results were compared to results from C57BL/6J (B6) age-matched control mice. Our results show that muscle healing upon cardiotoxin injury was accelerated in MRL/MpJ mice and characterized by reduced necrotic muscle area, reduced macrophage infiltration, and more regenerated myofibers (embryonic myosin heavy chain+/centronucleated fibers) at 3, 5, and 12 days postinjury, when compared to B6 age-matched control mice. These observations were associated with enhanced function of MPCs, including improved cell proliferation, differentiation, and resistance to stress, as well as increased muscle regenerative potential when compared to B6 MPCs. Mass spectrometry of serum proteins revealed higher levels of circulating antioxidants in MRL/MpJ mice when compared to B6 mice. Indeed, we found relatively higher gene expression of superoxide dismutase 1 (Sod1) and catalase (Cat) in MRL/MpJ MPCs. Depletion of Sod1 or Cat by small interfering RNA impaired myogenic potential of MRL/MpJ MPCs, indicating a role for these antioxidants in muscle repair. Taken together, these findings provide evidence that improved function of MPCs and higher levels of circulating antioxidants play important roles in accelerating muscle-healing capacity of MRL/MpJ mice. Stem Cells 2019;37:357-367.
Collapse
Affiliation(s)
- Chieh Tseng
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Krishna Sinha
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Haiying Pan
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yan Cui
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ping Guo
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Chih Yi Lin
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Fan Yang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Zhenhan Deng
- Department of Sports Medicine, Shenzhen Second People's Hospital, Shenzhen, Guangzhou, People's Republic of China
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aiping Lu
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| |
Collapse
|
39
|
Tasnim N, Chattopadhyay M, Joddar B. Scaffolds for tissue engineering of stomach. HANDBOOK OF TISSUE ENGINEERING SCAFFOLDS: VOLUME TWO 2019:633-646. [DOI: 10.1016/b978-0-08-102561-1.00025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
40
|
Masouminia M, Gelfand R, Kovanecz I, Vernet D, Tsao J, Salas R, Castro K, Loni L, Rajfer J, Gonzalez-Cadavid NF. Dyslipidemia Is a Major Factor in Stem Cell Damage Induced by Uncontrolled Long-Term Type 2 Diabetes and Obesity in the Rat, as Suggested by the Effects on Stem Cell Culture. J Sex Med 2018; 15:1678-1697. [PMID: 30527052 PMCID: PMC6645779 DOI: 10.1016/j.jsxm.2018.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 09/04/2018] [Accepted: 09/28/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Previous work showed that muscle-derived stem cells (MDSCs) exposed long-term to the milieu of uncontrolled type 2 diabetes (UC-T2D) in male obese Zucker (OZ) rats, were unable to correct the associated erectile dysfunction and the underlying histopathology when implanted into the corpora cavernosa, and were also imprinted with a noxious gene global transcriptional signature (gene-GTS), suggesting that this may interfere with their use as autografts in stem cell therapy. AIM To ascertain the respective contributions of dyslipidemia and hyperglycemia to this MDSC damage, clarify its mechanism, and design a bioassay to identify the damaged stem cells. METHODS Early diabetes MDSCs and late diabetes MDSCs were respectively isolated from nearly normal young OZ rats and moderately hyperglycemic and severely dyslipidemic/obese aged rats with erectile dysfunction. Monolayer cultures of early diabetic MDSCs were incubated 4 days in DMEM/10% fetal calf serum + or - aged OZ or lean Zucker serum from non-diabetic lean Zucker rats (0.5-5%) or with soluble palmitic acid (PA) (0.5-2 mM), cholesterol (CHOL) (50-400 mg/dL), or glucose (10-25 mM). MAIN OUTCOME MEASURE Fat infiltration was estimated by Oil red O, apoptosis by TUNEL, protein expression by Western blots, and gene-GTS and microRNA (miR)-GTS were determined in these stem cells' RNA. RESULTS Aged OZ serum caused fat infiltration, apoptosis, myostatin overexpression, and impaired differentiation. Some of these changes, and also a proliferation decrease occurred with PA and CHOL. The gene-GTS changes by OZ serum did not resemble the in vivo changes, but some occurred with PA and CHOL. The miR-GTS changes by OZ serum, PA, and CHOL resembled most of the in vivo changes. Hyperglycemia did not replicate most alterations. CLINICAL IMPLICATIONS MDSCs may be damaged in long-term UC-T2D/obese patients and be ineffective in autologous human stem cell therapy, which may be prevented by excluding the damaged MDSCs. STRENGTH & LIMITATIONS The in vitro test of MDSCs is innovative and fast to define dyslipidemic factors inducing stem cell damage, its mechanism, prevention, and counteraction. Confirmation is required in other T2D/obesity rat models and stem cells (including human), as well as miR-GTS biomarker validation as a stem cell damage biomarker. CONCLUSION Serum from long-term UC-T2D/obese rats or dyslipidemic factors induces a noxious phenotype and miR-GTS on normal MDSCs, which may lead in vivo to the repair inefficacy of late diabetic MDSCs. This suggests that autograft therapy with MDSCs in long-term UT-T2D obese patients may be ineffective, albeit this may be predictable by prior stem cell miR-GTS tests. Masouminia M, Gelfand R, Kovanecz I, et al. Dyslipidemia Is a Major Factor in Stem Cell Damage Induced by Uncontrolled Long-Term Type 2 Diabetes and Obesity in the Rat, as Suggested by the Effects on Stem Cell Culture. J Sex Med 2018;15:1678-1697.
Collapse
Affiliation(s)
- Maryam Masouminia
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Robert Gelfand
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Istvan Kovanecz
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA; Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Dolores Vernet
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - James Tsao
- Department of Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA
| | - Ruben Salas
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Kenny Castro
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Leila Loni
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Jacob Rajfer
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA
| | - Nestor F Gonzalez-Cadavid
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA, USA; Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA; Department of Medicine, Charles Drew University of Medicine and Science, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Xu Z, Yu L, Lu H, Feng W, Chen L, Zhou J, Yang X, Qi Z. A modified preplate technique for efficient isolation and proliferation of mice muscle-derived stem cells. Cytotechnology 2018; 70:1671-1683. [PMID: 30417280 DOI: 10.1007/s10616-018-0262-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/24/2018] [Indexed: 12/24/2022] Open
Abstract
We modified an existing protocol to develop a more efficient method to acquire and culture muscle-derived stem cells (MDSCs) and compared the characteristics of cells obtained from the two methods. This method is based on currently used multistep enzymatic digestion and preplate technique. During the replating process, we replaced the traditional medium with isolation medium to promote fibroblast-like cell adherence at initial replating step, which shortened the purifying duration by up to 4 days. Moreover, we modified the culture container to provide a stable microenvironment that promotes MDSC adherence. We compared the cell morphology, growth curve and the expression of specific markers (Sca-1, CD34, PAX7 and Desmin) between the two cell groups separately obtained from the two methods. Afterwards, we compared the neural differentiation capacity of MDSCs with other muscle-derived cell lineages. The protocol developed here is a fast and effective method to harvest and purify MDSCs from mice limb skeletal muscle.
Collapse
Affiliation(s)
- Zhuqiu Xu
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China
| | - Lu Yu
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China
| | - Haibin Lu
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China
| | - Weifeng Feng
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China
| | - Lulu Chen
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China
| | - Jing Zhou
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China
| | - Xiaonan Yang
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China.
| | - Zuoliang Qi
- Chinese Academy of Medical Science, Peking Union Medical College, Plastic Surgery Hospital, Beijing, 100041, China.
| |
Collapse
|
42
|
Burdzinska A, Dybowski B, Zarychta-Wiśniewska W, Kulesza A, Butrym M, Zagozdzon R, Graczyk-Jarzynka A, Radziszewski P, Gajewski Z, Paczek L. Intraurethral co-transplantation of bone marrow mesenchymal stem cells and muscle-derived cells improves the urethral closure. Stem Cell Res Ther 2018; 9:239. [PMID: 30241573 PMCID: PMC6151032 DOI: 10.1186/s13287-018-0990-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/16/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023] Open
Abstract
Background Cell therapy constitutes an attractive alternative to treat stress urinary incontinence. Although promising results have been demonstrated in this field, the procedure requires further optimization. The most commonly proposed cell types for intraurethral injections are muscle derived cells (MDCs) and mesenchymal stem/stromal cell (MSCs). The aim of this study was to evaluate the effects of MDC-MSC co-transplantation into the urethra. Methods Autologous transplantation of labeled MDCs, bone marrow MSCs or co-transplantation of MDC-MSC were performed in aged multiparous female goats (n = 6 in each group). The mean number of cells injected per animal was 29.6 × 106(± 4.3 × 106). PBS-injected animals constituted the control group (n = 5). Each animal underwent urethral pressure profile (UPP) measurements before and after the injection procedure. The maximal urethral closure pressure (MUCP) and functional area (FA) of UPPs were calculated. The urethras were collected at the 28th or the 84th day after transplantation. The marker fluorochrome (DID) was visualized and quantified using in vivo imaging system in whole explants. Myogenic differentiation of the graft was immunohistochemically evaluated. Results The grafted cells were identified in all urethras collected at day 28 regardless of injected cell type. At this time point the strongest DID-derived signal (normalized to the number of injected cells) was noted in the co-transplanted group. There was a distinct decline in signal intensity between day 28 and day 84 in all types of transplantation. Both MSCs and MDCs contributed to striated muscle formation if transplanted directly to the external urethral sphincter. In the MSC group those events were rare. If cells were injected into the submucosal region they remained undifferentiated usually packed in clearly distinguishable depots. The mean increase in MUCP after transplantation in comparison to the pre-transplantation state in the MDC, MSC and MDC-MSC groups was 12.3% (± 11.2%, not significant (ns)), 8.2% (± 9.6%, ns) and 24.1% (± 3.1%, p = 0.02), respectively. The mean increase in FA after transplantation in the MDC, MSC and MDC-MSC groups amounted to 17.8% (± 15.4%, ns), 15.2% (± 12.9%, ns) and 17.8% (± 2.5%, p = 0.04), respectively. Conclusions The results suggest that MDC-MSC co-transplantation provides a greater chance of improvement in urethral closure than transplantation of each population alone.
Collapse
Affiliation(s)
- Anna Burdzinska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland
| | - Bartosz Dybowski
- Department of Urology, Medical University of Warsaw, Warsaw, Poland
| | - Weronika Zarychta-Wiśniewska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland
| | - Agnieszka Kulesza
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland
| | - Marta Butrym
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland.,Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland.,Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Zdzislaw Gajewski
- Department of Large Animal Diseases with Clinic, Veterinary Research Centre and Center for Biomedical Research, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS - SGGW), Warsaw, Poland
| | - Leszek Paczek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland. .,Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
43
|
Lan KC, Wang CC, Yen YP, Yang RS, Liu SH, Chan DC. Islet-like clusters derived from skeletal muscle-derived stem/progenitor cells for autologous transplantation to control type 1 diabetes in mice. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S328-S335. [PMID: 30032651 DOI: 10.1080/21691401.2018.1492421] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A population of muscle-derived stem/progenitor cells (MDSPCs) contained in skeletal muscle is responsible for muscle regeneration. MDSPCs from mouse muscle have been shown to be capable of differentiating into pancreatic islet-like cells. However, the potency of MDSPCs to differentiate into functional islet-like cluster remains to be confirmed. The therapeutic potential of autologous MDSPCs transplantation on type 1 diabetes still remains unclear. Here, we investigated a four-stage method to induce the differentiation of MDSPCs into insulin-producing clusters in vitro, and tested the autologous transplantation to control type 1 diabetes in mice. MDSPCs isolated from the skeletal muscles of mice possessed the ability to form islet-like clusters through several stages of differentiation. The expressions of pancreatic progenitor-related genes, insulin, and islet-related genes were significantly upregulated in islet-like clusters determined by the quantitative reverse transcription polymerase chain reaction. The autologous islet-like clusters transplantation effectively improved hyperglycaemia and glucose intolerance and increased the survival rate in streptozotocin-induced diabetic mice without the use of immunosuppressants. Taken together, these results provide evidence that MDSPCs from murine muscle tissues are capable of differentiating into insulin-producing clusters, which possess insulin-producing ability in vitro and in vivo, and have the potential for autologous transplantation to control type 1 diabetes.
Collapse
Affiliation(s)
- Kuo-Cheng Lan
- a Department of Emergency Medicine , Tri-Service General Hospital, National Defense Medical Center , Taipei , Taiwan
| | - Ching-Chia Wang
- b Department of Pediatrics , College of Medicine, National Taiwan University , Taipei , Taiwan
| | - Yuan-Peng Yen
- c College of Medicine , Institute of Toxicology, National Taiwan University , Taipei , Taiwan
| | - Rong-Sen Yang
- d Department of Orthopaedics, College of Medicine , National Taiwan University , Taipei , Taiwan
| | - Shing-Hwa Liu
- b Department of Pediatrics , College of Medicine, National Taiwan University , Taipei , Taiwan.,c College of Medicine , Institute of Toxicology, National Taiwan University , Taipei , Taiwan.,e Department of Medical Research , China Medical University Hospital, China Medical University , Taichung , Taiwan
| | - Ding-Cheng Chan
- f Department of Geriatrics and Gerontology , National Taiwan University , Taipei , Taiwan
| |
Collapse
|
44
|
Wang J, Zhang F, Yang H, Wu H, Cui R, Zhao Y, Jiao C, Wang X, Liu X, Wu L, Li G, Wu X. Effect of TEAD4 on multilineage differentiation of muscle-derived stem cells. Am J Transl Res 2018; 10:998-1011. [PMID: 29636889 PMCID: PMC5883140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/10/2018] [Indexed: 06/08/2023]
Abstract
TEAD4 is a member of transcriptional enhancer factor (TEF) family of transcription factors and plays a pivotal role in regulating embryonic development and muscle regeneration. Known previously, dysfunction of TEAD4 in mouse myoblasts impairs myotube development. However, the effects of TEAD4 on multipotency of muscle-derived stem cells (MDSCs) have not been clearly understood. Recently, bovine MDSCs (bMDSCs) were successfully isolated from adult bovine muscle. Our derived bMDSCs could differentiate into mesodermal cells, including myotubes, adipocytes, and osteoid cells. Our results also revealed that bMDSCs had the capacity to develop into ectodermal and endodermal lineages including neuron-like cells and insulin-secreting cells. After TEAD4 knock-down (TEAD4-KD), bMDSCs still kept the original capacity to differentiate into neuron-like cells and insulin-secreting cells, as shown by acquisition of both neuronal and pancreatic markers normally expressed in differentiated cells. However, up-regulation of CAV3 and βMHC failed during myogenesis of bMDSCs with TEAD4-KD, although TEAD4-KD in bMDSCs did not affect osteoid cells and myotube formation. More interestingly, adipogenic differentiation of TEAD4-KD bMDSCs was significantly suppressed. During adipogenic differentiation, TEAD4-KD systematically impaired upregulation of TEAD1, TEAD2, and TEAD3, as well as the activation of C/EBP2, ADD1, and PPARγ as the key transcription factors for adipogenic differentiation. Finally, TEAD4-KD led to the failure of adipogenesis from bMDSCs. Together, our results support that TEAD4 is essential during adipogenic differentiation of bMDSCs. It has little effect on myogenesis of bMDSCs, and does not affect ostegenesis, neurogenesis, or pancreatic differentiation of bMDSCs. Our findings will be helpful for future study on the roles of the TEAD family during differentiation of MDSCs, and for controlling MDSC differentiation for stem cell applications.
Collapse
Affiliation(s)
- Jinze Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Feixu Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Huidi Yang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
- School of Basic Medical Sciences, Inner Mongolia Medical CollegeHohhot 010110, People’s Republic of China
| | - Huikuan Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Rong Cui
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Yunjie Zhao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Cuihua Jiao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Xianxin Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Xin Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Liqiong Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Guangpeng Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| | - Xia Wu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia UniversityHohhot 010070, Inner Mongolia, People’s Republic of China
| |
Collapse
|
45
|
Matthias N, Hunt SD, Wu J, Lo J, Smith Callahan LA, Li Y, Huard J, Darabi R. Volumetric muscle loss injury repair using in situ fibrin gel cast seeded with muscle-derived stem cells (MDSCs). Stem Cell Res 2018; 27:65-73. [PMID: 29331939 PMCID: PMC5851454 DOI: 10.1016/j.scr.2018.01.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/02/2017] [Accepted: 01/05/2018] [Indexed: 12/22/2022] Open
Abstract
Volumetric muscle defect, caused by trauma or combat injuries, is a major health concern leading to severe morbidity. It is characterized by partial or full thickness loss of muscle and its bio-scaffold, resulting in extensive fibrosis and scar formation. Therefore, the ideal therapeutic option is to use stem cells combined with bio-scaffolds to restore muscle. For this purpose, muscle-derived stem cells (MDSCs) are a great candidate due to their unique multi-lineage differentiation potential. In this study, we evaluated the regeneration potential of MDSCs for muscle loss repair using a novel in situ fibrin gel casting. Muscle defect was created by a partial thickness wedge resection in the tibialis anterior (TA)muscles of NSG mice which created an average of 25% mass loss. If untreated, this defect leads to severe muscle fibrosis. Next, MDSCs were delivered using a novel in situ fibrin gel casting method. Our results demonstrated MDSCs are able to engraft and form new myofibers in the defect when casted along with fibrin gel. LacZ labeled MDSCs were able to differentiate efficiently into new myofibers and significantly increase muscle mass. This was also accompanied by significant reduction of fibrotic tissue in the engrafted muscles. Furthermore, transplanted cells also contributed to new vessel formation and satellite cell seeding. These results confirmed the therapeutic potential of MDSCs and feasibility of direct in situ casting of fibrin/MDSC mixture to repair muscle mass defects.
Collapse
Affiliation(s)
- Nadine Matthias
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Samuel D Hunt
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Jianbo Wu
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Jonathan Lo
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States
| | - Laura A Smith Callahan
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States; The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States; Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States; Department of Nanomedicine and Biomedical Engineering, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Yong Li
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States; Department of Pediatric Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Johnny Huard
- Department of Orthopedic Surgery, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Radbod Darabi
- Center for Stem Cell and Regenerative Medicine (CSCRM) and the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), United States; The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States.
| |
Collapse
|
46
|
Wang H, Yi J, Li X, Xiao Y, Dhakal K, Zhou J. ALS-associated mutation SOD1 G93A leads to abnormal mitochondrial dynamics in osteocytes. Bone 2018; 106:126-138. [PMID: 29030231 PMCID: PMC5718158 DOI: 10.1016/j.bone.2017.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022]
Abstract
While the death of motor neuron is a pathological hallmark of amyotrophic lateral sclerosis (ALS), defects in other cell types or organs may also actively contribute to ALS disease progression. ALS patients experience progressive skeletal muscle wasting that may not only exacerbate neuronal degeneration, but likely has a significant impact on bone function. In our previous published study, we have discovered severe bone loss in an ALS mouse model with overexpression of ALS-associated mutation SOD1G93A (G93A). Here we further provide a mechanistic understanding of the bone loss in ALS animal and cellular models. Combining mitochondrial fluorescent indicators and confocal live cell imaging, we discovered abnormalities in mitochondrial network and dynamics in primary osteocytes derived from the same ALS mouse model G93A. Those mitochondrial defects occur in ALS mice after the onset of neuromuscular symptoms, indicating that mitochondria in bone cells respond to muscle atrophy during ALS disease progression. To examine whether ALS mutation has a direct contribution to mitochondrial dysfunction independent of muscle atrophy, we evaluated mitochondrial morphology and motility in cultured osteocytes (MLO-Y4) with overexpression of mitochondrial targeted SOD1G93A. Compared with osteocytes overexpressing the wild type SOD1 as a control, the SOD1G93A osteocytes showed similar defects in mitochondrial network and dynamic as that of the primary osteocytes derived from the ALS mouse model. In addition, we further discovered that overexpression of SOD1G93A enhanced the expression level of dynamin-related protein 1 (Drp1), a key protein promoting mitochondrial fission activity, and reduced the expression level of optic atrophy protein 1 (OPA1), a key protein related to mitochondrial fusion. A specific mitochondrial fission inhibitor (Mdivi-1) partially reversed the effect of SOD1G93A on mitochondrial network and dynamics, indicating that SOD1G93A likely promotes mitochondrial fission, but suppresses the fusion activity. Our data provide the first evidence that mitochondria show abnormality in osteocytes derived from an ALS mouse model. The accumulation of mutant SOD1G93A protein inside mitochondria directly causes dysfunction in mitochondrial dynamics in cultured MLO-Y4 osteocytes. In addition, the ALS mutation SOD1G93A-mediated dysfunction in mitochondrial dynamics is associated with an enhanced apoptosis in osteocytes, which could be a potential mechanism underlying the bone loss during ALS progression.
Collapse
Affiliation(s)
- Huan Wang
- Kansas City University of Medicine and Bioscience, Kansas City, MO, USA
| | - Jianxun Yi
- Kansas City University of Medicine and Bioscience, Kansas City, MO, USA
| | - Xuejun Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO, USA
| | - Yajuan Xiao
- Kansas City University of Medicine and Bioscience, Kansas City, MO, USA
| | - Kamal Dhakal
- Kansas City University of Medicine and Bioscience, Kansas City, MO, USA
| | - Jingsong Zhou
- Kansas City University of Medicine and Bioscience, Kansas City, MO, USA.
| |
Collapse
|
47
|
Shenoy P S, Bose B. Hepatic perivascular mesenchymal stem cells with myogenic properties. J Tissue Eng Regen Med 2017. [PMID: 28627746 DOI: 10.1002/term.2503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Pericytes are multipotent mesenchymal stem cells located on the walls of blood vessels in various organs and are characterized as CD146+ cells. In this study, we first immunohistochemically detected pericytes in the perivascular regions of liver from two mouse genotypes, namely wild-type (WT) and myostatin null (Mstn-/- ). We further isolated pericytes using sorting as CD146+ CD34- CD56- CD45- cells. The main finding of this study involves the contrasting fibrogenic vs. myogenic behaviour of liver pericytes from WT and Mstn-/- mice, respectively. Sorted CD146+ liver pericytes (WT and Mstn-/- ) expressed PDGFRβ, NG2, vimentin, adult stem cell markers CD73, CD105, CD44 and could be readily differentiated into adipogenic, osteogenic and chondrogenic lineages. Furthermore, these CD146+ cells from WT and Mstn-/- livers did not express myostatin, in contrast to the total liver tissue of WT. The absence of αSMA and GFAP made these cells easily distinguishable from hepatic stellate cells. When subjected to standard myogenic differentiation with low serum the CD146+ cells from WT liver differentiated into myofibroblasts (fibrogenic) and the CD146+ cells from Mstn-/- liver differentiated into multinucleated myotubes (myogenic). Finally, we transplanted CD146+ pericytes into tibialis anterior muscle of dystrophic mice and established the generation of novel myofibres, thereby proving their cell therapy potential. The liver tissue microenvironment with myostatin in WT and the absence of myostatin in Mstn-/- conditions might exert a paracrine effect in determining the fate of pericyte-like cells in the liver.
Collapse
Affiliation(s)
- Sudheer Shenoy P
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Bipasha Bose
- School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
48
|
Persichini T, Funari A, Colasanti M, Sacchetti B. Clonogenic, myogenic progenitors expressing MCAM/CD146 are incorporated as adventitial reticular cells in the microvascular compartment of human post-natal skeletal muscle. PLoS One 2017; 12:e0188844. [PMID: 29186180 PMCID: PMC5706678 DOI: 10.1371/journal.pone.0188844] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/14/2017] [Indexed: 12/13/2022] Open
Abstract
Recent observation identifies subendothelial (mural) cells expressing MCAM, a specific system of clonogenic, self-renewing, osteoprogenitors (a.k.a, "mesenchymal stem cells") in the microvascular compartment of post-natal human bone marrow (BM). In this study, we used MCAM/CD146, as a marker to localize, isolate and assay subendothelial clonogenic cells from the microvasculature of postnatal human skeletal muscle. We show here that these cells share with their BM counterpart, anatomic position (subendothelial/adventitial) and ex vivo clonogenicity (CFU-Fs). When assayed under the stringent conditions, these cells display a high spontaneous myogenic potential (independent of co-culture with myoblasts or of in vivo fusion with local myoblasts), which is otherwise only attained in cultures of satellite cells. These muscle-derived mural cells activated a myogenic program in culture. Cultured CD146+ cells expressed the myogenic factors (Pax7, Pax3 and Myf5), NCAM/CD56, desmin as well as proteins characteristic of more advanced myogenic differentiation, such as myosin heavy chain. In vivo, these cells spontaneously generate myotubes and myofibrils. These data identify the anatomy and phenotype of a novel class of committed myogenic progenitor in human post-natal skeletal muscle of subendothelial cells associated with the abluminal surface of microvascular compartment distinct from satellite cells.
Collapse
Affiliation(s)
| | - Alessia Funari
- Department of Molecular Medicine, University “Sapienza”, Rome, Italy
| | | | - Benedetto Sacchetti
- Department of Science, University ROMA TRE, Rome, Italy
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
49
|
Stilhano RS, Samoto VY, Silva LM, Pereira GJ, Erustes AG, Smaili SS, Won Han S. Reduction in skeletal muscle fibrosis of spontaneously hypertensive rats after laceration by microRNA targeting angiotensin II receptor. PLoS One 2017; 12:e0186719. [PMID: 29059221 PMCID: PMC5653346 DOI: 10.1371/journal.pone.0186719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/08/2017] [Indexed: 12/23/2022] Open
Abstract
Regeneration of injured skeletal muscles is affected by fibrosis, which can be improved by the administration of angiotensin II (AngII) receptor (ATR) blockers in normotensive animals. However, the role of ATR in skeletal muscle fibrosis in hypertensive organisms has not been investigated yet. The tibialis anterior (TA) muscle of spontaneously hypertensive (SHR) and Wistar rats (WR) were lacerated and a lentivector encoding a microRNA targeting AngII receptor type 1 (At1) (Lv-mirAT1a) or control (Lv-mirCTL) was injected. The TA muscles were collected after 30 days to evaluate fibrosis by histology and gene expression by real-time quantitative PCR (RT-qPCR) and Western blot. SHR's myoblasts were analyzed by RT-qPCR, 48 h after transduction. In the SHR's TA, AT1 protein expression was 23.5-fold higher than in WR without injury, but no difference was observed in the angiotensin II receptor type 2 (AT2) protein expression. TA laceration followed by suture (LS) produced fibrosis in the SHR (23.3±8.5%) and WR (7.9±1.5%). Lv-mirAT1 treatment decreased At1 gene expression in 50% and reduced fibrosis to 7% 30 days after. RT-qPCR showed that reduction in At1 expression is due to downregulation of the At1a but not of the At1b. RT-qPCR of myoblasts from SHR transduced with Lv-mirAT1a showed downregulation of the Tgf-b1, Tgf-b2, Smad3, Col1a1, and Col3a1 genes by mirAT1a. In vivo and in vitro studies indicate that hypertension overproduces skeletal muscle fibrosis, and AngII-AT1a signaling is the main pathway of fibrosis in SHR. Moreover, muscle fibrosis can be treated specifically by in loco injection of Lv-mirAT1a without affecting other organs.
Collapse
Affiliation(s)
- Roberta Sessa Stilhano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Vivian Yochiko Samoto
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Leonardo Martins Silva
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Gustavo José Pereira
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Adolfo Garcia Erustes
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Soraya Soubhi Smaili
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
| | - Sang Won Han
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
50
|
Rajabi S, Jalili-Firoozinezhad S, Ashtiani MK, Le Carrou G, Tajbakhsh S, Baharvand H. Effect of chemical immobilization of SDF-1α into muscle-derived scaffolds on angiogenesis and muscle progenitor recruitment. J Tissue Eng Regen Med 2017; 12:e438-e450. [DOI: 10.1002/term.2479] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/04/2017] [Accepted: 05/09/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Sarah Rajabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
| | - Sasan Jalili-Firoozinezhad
- Department of Cell Engineering, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
- Departments of Biomedicine and Surgery; University Hospital Basel; Basel Switzerland
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
| | - Gilles Le Carrou
- Department of Developmental and Stem Cell Biology, Stem Cells and Development Unit, CNRS URA 3738; Institut Pasteur Paris; Paris France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Stem Cells and Development Unit, CNRS URA 3738; Institut Pasteur Paris; Paris France
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology, ACECR; Tehran Iran
- Department of Developmental Biology; University of Science and Culture; Tehran Iran
| |
Collapse
|