1
|
Lin W, Chow SKH, Cui C, Liu C, Wang Q, Chai S, Wong RMY, Zhang N, Cheung WH. Wnt/β-catenin signaling pathway as an important mediator in muscle and bone crosstalk: A systematic review. J Orthop Translat 2024; 47:63-73. [PMID: 39007034 PMCID: PMC11245956 DOI: 10.1016/j.jot.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/21/2024] [Accepted: 06/02/2024] [Indexed: 07/16/2024] Open
Abstract
Background The interaction between muscle and bone is shown to be clinically important but the underlying mechanisms are largely unknown. The canonical Wnt/β-catenin signaling pathway is reported to be involved in muscle-bone crosstalk, but its detailed function remains unclear. This systematic review aims to investigate and elucidate the role of the Wnt/β-catenin signaling pathways in muscle-bone crosstalk. Methods We conducted a literature search on the Web of Science, PubMed, EBSCO and Embase with keywords "Wnt*", "bone*" and "muscle*". A systematic review was completed according to the guideline of preferred reporting items of systematic reviews and meta-analyses (PRISMA). Data synthesis included species (human, animal or cell type used), treatments involved, outcome measures and key findings with respect to Wnts. Results Seventeen papers were published from 2007 to 2021 and were extracted from a total of 1529 search results in the databases of Web of Science (468 papers), PubMed (457 papers), EBSCO (371) and Embase (233). 12 Wnt family members were investigated in the papers, including Wnt1, Wnt2, Wnt2b, Wnt3a, Wnt4, Wnt5a, Wnt8a, Wnt8b, Wnt9a, Wnt10a, Wnt10b and Wnt16. Many studies showed that muscles were able to increase or decrease osteogenesis of bone, while bone increased myogenesis of muscle through Wnt/β-catenin signaling pathways. Wnt3a, Wnt4 and Wnt10b were shown to play important roles in the crosstalk between muscle and bone. Conclusions Wnt3a, Wnt4 and Wnt10b are found to play important mediatory roles in muscle-bone crosstalk. The role of Wnt4 was mostly found to regulate muscle from the bone side. Whilst the role of Wnt10b during muscle ageing was proposed, current evidence is insufficient to clarify the specific role of Wnt/β-catenin signaling in the interplay between sarcopenia and osteoporosis. More future studies are required to investigate the exact regulatory roles of Wnts in muscle-bone crosstalk in musculoskeletal disease models such as sarcopenia and osteoporosis. Translational potential of this article The systematic review provides an extensive overview to reveal the roles of Wnt/β-catenin signaling pathways in muscle-bone crosstalk. These results provide novel research directions to further understand the underlying mechanism of sarcopenia, osteoporosis, and their crosstalk, finally helping the future development of new therapeutic interventions.
Collapse
Affiliation(s)
- Wujian Lin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Department of Rehabilitation Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Simon Kwoon Ho Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, USA
| | - Can Cui
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Chaoran Liu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Qianjin Wang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Senlin Chai
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Ronald Man Yeung Wong
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Ning Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Wing Hoi Cheung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| |
Collapse
|
2
|
Shin DI, Jin YJ, Noh S, Yun HW, Park DY, Min BH. Exosomes Secreted During Myogenic Differentiation of Human Fetal Cartilage-Derived Progenitor Cells Promote Skeletal Muscle Regeneration through miR-145-5p. Tissue Eng Regen Med 2024; 21:487-497. [PMID: 38294592 PMCID: PMC10987463 DOI: 10.1007/s13770-023-00618-w] [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: 11/03/2023] [Revised: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Currently, there is no apparent treatment for sarcopenia, which is characterized by diminished myoblast function. We aimed to manufacture exosomes that retain the myogenic differentiation capacity of human fetal cartilage-derived progenitor cells (hFCPCs) and investigate their muscle regenerative efficacy in myoblasts and a sarcopenia rat model. METHODS The muscle regeneration potential of exosomes (F-Exo) secreted during myogenic differentiation of hFCPCs was compared to human bone marrow mesenchymal stem cells-derived (hBMSCs) exosomes (B-Exo) in myoblasts and sarcopenia rat model. The effect of F-Exo was analyzed through known microRNAs (miRNAs) analysis. The mechanism of action of F-Exo was confirmed by measuring the expression of proteins involved in the Wnt signaling pathway. RESULTS F-Exo and B-Exo showed similar exosome characteristics. However, F-Exo induced the expression of muscle markers (MyoD, MyoG, and MyHC) and myotube formation in myoblasts more effectively than B-Exo. Moreover, F-Exo induced greater increases in muscle fiber cross-sectional area and muscle mass compared to B-Exo in a sarcopenia rat. The miR-145-5p, relevant to muscle regeneration, was found in high concentrations in the F-Exo, and RNase pretreatment reduced the efficacy of exosomes. The effects of F-Exo on the expression of myogenic markers in myoblasts were paralleled by the miR-145-5p mimics, while the inhibitor partially negated this effect. F-Exo was involved in the Wnt signaling pathway by enhancing the expression of Wnt5a and β-catenin. CONCLUSION F-Exo improved muscle regeneration by activating the Wnt signaling pathway via abundant miR-145-5p, mimicking the remarkable myogenic differentiation potential of hFCPCs.
Collapse
Affiliation(s)
- Dong Il Shin
- Department of Molecular Science and Technology, Ajou University Graduate School, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Yong Jun Jin
- Cell Therapy Center, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
- Department of Orthopedic Surgery, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Sujin Noh
- Cell Therapy Center, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Hee-Woong Yun
- Cell Therapy Center, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
- Department of Orthopedic Surgery, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Do Young Park
- Cell Therapy Center, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
- Department of Orthopedic Surgery, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Byoung-Hyun Min
- Department of Molecular Science and Technology, Ajou University Graduate School, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea.
- Cell Therapy Center, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea.
- Department of Orthopedic Surgery, Ajou University School of Medicine, 206 Worldcup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea.
| |
Collapse
|
3
|
Naraoka Y, Mabuchi Y, Kiuchi M, Kumagai K, Hisamatsu D, Yoneyama Y, Takebe T, Akazawa C. Quality Control of Stem Cell-Based Cultured Meat According to Specific Differentiation Abilities. Cells 2024; 13:135. [PMID: 38247826 PMCID: PMC10814720 DOI: 10.3390/cells13020135] [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: 11/06/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
The demand for stem cell-based cultured meat as an alternative protein source is increasing in response to global food scarcity. However, the definition of quality controls, including appropriate growth factors and cell characteristics, remains incomplete. Cluster of differentiation (CD) 29 is ubiquitously expressed in bovine muscle tissue and is a marker of progenitor cells in cultured meat. However, CD29+ cells are naturally heterogeneous, and this quality control issue must be resolved. In this study, the aim was to identify the subpopulation of the CD29+ cell population with potential utility in cultured meat production. The CD29+ cell population exhibited heterogeneity, discernible through the CD44 and CD344 markers. CD29+CD44-CD344- cells displayed the ability for long-term culture, demonstrating high adipogenic potential and substantial lipid droplet accumulation, even within 3D cultures. Conversely, CD29+CD44+ cells exhibited rapid proliferation but were not viable for prolonged culture. Using cells suitable for adipocyte and muscle differentiation, we successfully designed meat buds, especially those rich in fat. Collectively, the identification and comprehension of distinct cell populations within bovine tissues contribute to quality control predictions in meat production. They also aid in establishing a stable and reliable cultured meat production technique.
Collapse
Grants
- JPMJMI18CB Japan Science and Technology Agency
- JP21H03328 Ministry of Education, Culture, Sports, Science and Technology
- JP19K19986 Ministry of Education, Culture, Sports, Science and Technology
- JP22K17699 Ministry of Education, Culture, Sports, Science and Technology
- no number Otsuka Holdings Co., Ltd.
Collapse
Affiliation(s)
- Yuna Naraoka
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.N.); (Y.M.); (M.K.); (K.K.); (D.H.)
| | - Yo Mabuchi
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.N.); (Y.M.); (M.K.); (K.K.); (D.H.)
- Department of Clinical Regenerative Medicine, Fujita Medical Innovation Center, Fujita Health University, 1-1-4, Hanedakuko, Ota-ku, Tokyo 144-0041, Japan
| | - Mai Kiuchi
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.N.); (Y.M.); (M.K.); (K.K.); (D.H.)
| | - Kyoko Kumagai
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.N.); (Y.M.); (M.K.); (K.K.); (D.H.)
| | - Daisuke Hisamatsu
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.N.); (Y.M.); (M.K.); (K.K.); (D.H.)
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (Y.Y.); (T.T.)
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; (Y.Y.); (T.T.)
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Division of Developmental Biology, Cincinnati Children Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children Hospital Medical Center, Cincinnati, OH 45229-3039, USA
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chihiro Akazawa
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (Y.N.); (Y.M.); (M.K.); (K.K.); (D.H.)
| |
Collapse
|
4
|
Ochi E, Barrington A, Wehling‐Henricks M, Avila M, Kuro‐o M, Tidball JG. Klotho regulates the myogenic response of muscle to mechanical loading and exercise. Exp Physiol 2023; 108:1531-1547. [PMID: 37864311 PMCID: PMC10841225 DOI: 10.1113/ep091263] [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: 04/14/2023] [Accepted: 08/16/2023] [Indexed: 10/22/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does the hormone Klotho affect the myogenic response of muscle cells to mechanical loading or exercise? What is the main finding and its importance? Klotho prevents direct, mechanical activation of genes that regulate muscle differentiation, including genes that encode the myogenic regulatory factor myogenin and proteins in the canonical Wnt signalling pathway. Similarly, elevated levels of klotho expression in vivo prevent the exercise-induced increase in myogenin-expressing cells and reduce exercise-induced activation of the Wnt pathway. These findings demonstrate a new mechanism through which the responses of muscle to the mechanical environment are regulated. ABSTRACT Muscle growth is influenced by changes in the mechanical environment that affect the expression of genes that regulate myogenesis. We tested whether the hormone Klotho could influence the response of muscle to mechanical loading. Applying mechanical loads to myoblasts in vitro increased RNA encoding transcription factors that are expressed in activated myoblasts (Myod) and in myogenic cells that have initiated terminal differentiation (Myog). However, application of Klotho to myoblasts prevented the loading-induced activation of Myog without affecting loading-induced activation of Myod. This indicates that elevated Klotho inhibits mechanically-induced differentiation of myogenic cells. Elevated Klotho also reduced the transcription of genes encoding proteins involved in the canonical Wnt pathway or their target genes (Wnt9a, Wnt10a, Ccnd1). Because the canonical Wnt pathway promotes differentiation of myogenic cells, these findings indicate that Klotho inhibits the differentiation of myogenic cells experiencing mechanical loading. We then tested whether these effects of Klotho occurred in muscles of mice experiencing high-intensity interval training (HIIT) by comparing wild-type mice and klotho transgenic mice. The expression of a klotho transgene combined with HIIT synergized to tremendously elevate numbers of Pax7+ satellite cells and activated MyoD+ cells. However, transgene expression prevented the increase in myogenin+ cells caused by HIIT in wild-type mice. Furthermore, transgene expression diminished the HIIT-induced activation of the canonical Wnt pathway in Pax7+ satellite cells. Collectively, these findings show that Klotho inhibits loading- or exercise-induced activation of muscle differentiation and indicate a new mechanism through which the responses of muscle to the mechanical environment are regulated.
Collapse
Affiliation(s)
- Eisuke Ochi
- Faculty of Bioscience and Applied ChemistryHosei UniversityTokyoJapan
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Alice Barrington
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | | | - Marcus Avila
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Makoto Kuro‐o
- Division of Anti‐Aging MedicineCenter for Molecular MedicineJichi Medical UniversityTochigiJapan
| | - James G. Tidball
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular, Cellular & Integrative Physiology ProgramUniversity of CaliforniaLos AngelesCAUSA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCAUSA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
5
|
Oprescu SN, Baumann N, Chen X, Sun Q, Zhao Y, Yue F, Wang H, Kuang S. Sox11 is enriched in myogenic progenitors but dispensable for development and regeneration of the skeletal muscle. Skelet Muscle 2023; 13:15. [PMID: 37705115 PMCID: PMC10498607 DOI: 10.1186/s13395-023-00324-0] [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: 03/09/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Transcription factors (TFs) play key roles in regulating differentiation and function of stem cells, including muscle satellite cells (MuSCs), a resident stem cell population responsible for postnatal regeneration of the skeletal muscle. Sox11 belongs to the Sry-related HMG-box (SOX) family of TFs that play diverse roles in stem cell behavior and tissue specification. Analysis of single-cell RNA-sequencing (scRNA-seq) datasets identify a specific enrichment of Sox11 mRNA in differentiating but not quiescent MuSCs. Consistent with the scRNA-seq data, Sox11 levels increase during differentiation of murine primary myoblasts in vitro. scRNA-seq data comparing muscle regeneration in young and old mice further demonstrate that Sox11 expression is reduced in aged MuSCs. Age-related decline of Sox11 expression is associated with reduced chromatin contacts within the topologically associating domains. Unexpectedly, Myod1Cre-driven deletion of Sox11 in embryonic myoblasts has no effects on muscle development and growth, resulting in apparently healthy muscles that regenerate normally. Pax7CreER- or Rosa26CreER- driven (MuSC-specific or global) deletion of Sox11 in adult mice similarly has no effects on MuSC differentiation or muscle regeneration. These results identify Sox11 as a novel myogenic differentiation marker with reduced expression in quiescent and aged MuSCs, but the specific function of Sox11 in myogenesis remains to be elucidated.
Collapse
Affiliation(s)
- Stephanie N Oprescu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Nick Baumann
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiyue Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Qiang Sun
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong, China
| | - Yu Zhao
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong, China
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Huating Wang
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong, China
| | - Shihuan Kuang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
6
|
Kajabadi N, Low M, Jacques E, Lad H, Tung LW, Babaeijandaghi F, Gamu D, Zelada D, Wong CK, Chang C, Yi L, Wosczyna MN, Rando TA, Henríquez JP, Gibson WT, Gilbert PM, Rossi FMV. Activation of β-catenin in mesenchymal progenitors leads to muscle mass loss. Dev Cell 2023; 58:489-505.e7. [PMID: 36898377 DOI: 10.1016/j.devcel.2023.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/12/2022] [Accepted: 02/10/2023] [Indexed: 03/11/2023]
Abstract
Loss of muscle mass is a common manifestation of chronic disease. We find the canonical Wnt pathway to be activated in mesenchymal progenitors (MPs) from cancer-induced cachectic mouse muscle. Next, we induce β-catenin transcriptional activity in murine MPs. As a result, we observe expansion of MPs in the absence of tissue damage, as well as rapid loss of muscle mass. Because MPs are present throughout the organism, we use spatially restricted CRE activation and show that the induction of tissue-resident MP activation is sufficient to induce muscle atrophy. We further identify increased expression of stromal NOGGIN and ACTIVIN-A as key drivers of atrophic processes in myofibers, and we verify their expression by MPs in cachectic muscle. Finally, we show that blocking ACTIVIN-A rescues the mass loss phenotype triggered by β-catenin activation in MPs, confirming its key functional role and strengthening the rationale for targeting this pathway in chronic disease.
Collapse
Affiliation(s)
- Nasim Kajabadi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Marcela Low
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Carrera de Química y Farmacia, Facultad de Medicina y Ciencia, Universidad San Sebastián, General Lagos 1163, 5090000 Valdivia, Chile
| | - Erik Jacques
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Heta Lad
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Farshad Babaeijandaghi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Daniel Gamu
- BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, C201, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Diego Zelada
- Neuromuscular Studies Laboratory (NeSt Lab), GDeP, Department of Cell Biology, Universidad de Concepción, Concepción, Chile
| | - Chi Kin Wong
- BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, C201, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Chihkai Chang
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lin Yi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Michael N Wosczyna
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Musculoskeletal Research Center, Bioengineering Institute, Department of Orthopedic Surgery, NYU Grossman School of Medicine, New York, NY 10010, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), GDeP, Department of Cell Biology, Universidad de Concepción, Concepción, Chile
| | - William T Gibson
- BC Children's Hospital Research Institute, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, C201, 4500 Oak Street, Vancouver, BC V6H 3N1, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
7
|
Balatskyi VV, Sowka A, Dobrzyn P, Piven OO. WNT/β-catenin pathway is a key regulator of cardiac function and energetic metabolism. Acta Physiol (Oxf) 2023; 237:e13912. [PMID: 36599355 DOI: 10.1111/apha.13912] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
The WNT/β-catenin pathway is a master regulator of cardiac development and growth, and its activity is low in healthy adult hearts. However, even this low activity is essential for maintaining normal heart function. Acute activation of the WNT/β-catenin signaling cascade is considered to be cardioprotective after infarction through the upregulation of prosurvival genes and reprogramming of metabolism. Chronically high WNT/β-catenin pathway activity causes profibrotic and hypertrophic effects in the adult heart. New data suggest more complex functions of β-catenin in metabolic maturation of the perinatal heart, establishing an adult pattern of glucose and fatty acid utilization. Additionally, low basal activity of the WNT/β-catenin cascade maintains oxidative metabolism in the adult heart, and this pathway is reactivated by physiological or pathological stimuli to meet the higher energy needs of the heart. This review summarizes the current state of knowledge of the organization of canonical WNT signaling and its function in cardiogenesis, heart maturation, adult heart function, and remodeling. We also discuss the role of the WNT/β-catenin pathway in cardiac glucose, lipid metabolism, and mitochondrial physiology.
Collapse
Affiliation(s)
- Volodymyr V Balatskyi
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Adrian Sowka
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Dobrzyn
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Oksana O Piven
- Laboratory of Molecular Medical Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Department of Human Genetics, Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
8
|
Cui Y, Yi Q, Sun W, Huang D, Zhang H, Duan L, Shang H, Wang D, Xiong J. Molecular basis and therapeutic potential of myostatin on bone formation and metabolism in orthopedic disease. Biofactors 2023; 49:21-31. [PMID: 32997846 DOI: 10.1002/biof.1675] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022]
Abstract
Myostatin, a member of the transforming growth factor-β (TGF-β) superfamily, is a key autocrine/paracrine inhibitor of skeletal muscle growth. Recently, researchers have postulated that myostatin is a negative regulator of bone formation and metabolism. Reportedly, myostatin is highly expressed in the fracture area, affecting the endochondral ossification process during the early stages of fracture healing. Furthermore, myostatin is highly expressed in the synovium of patients with rheumatoid arthritis (RA) and is an effective therapeutic target for interfering with osteoclast formation and joint destruction in RA. Thus, myostatin is a potent anti-osteogenic factor and a direct modulator of osteoclast differentiation. Evaluation of the molecular pathway revealed that myostatin can activate SMAD and mitogen-activated protein kinase signaling pathways, inhibiting the Wnt/β-catenin pathway to synergistically regulate muscle and bone growth and metabolism. In summary, inhibition of myostatin or the myostatin signaling pathway has therapeutic potential in the treatment of orthopedic diseases. This review focused on the effects of myostatin on bone formation and metabolism and discussed the potential therapeutic effects of inhibiting myostatin and its pathways in related orthopedic diseases.
Collapse
Affiliation(s)
- Yinxing Cui
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Qian Yi
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Weichao Sun
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Dixi Huang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Hui Zhang
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
- University of South China, Hengyang, Hunan, China
| | - Li Duan
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Hongxi Shang
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Daping Wang
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| | - Jianyi Xiong
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Department of Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, Guangdong, China
| |
Collapse
|
9
|
Macrophages and Wnts in Tissue Injury and Repair. Cells 2022; 11:cells11223592. [PMID: 36429021 PMCID: PMC9688352 DOI: 10.3390/cells11223592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are important players in the immune system that sense various tissue challenges and trigger inflammation. Tissue injuries are followed by inflammation, which is tightly coordinated with tissue repair processes. Dysregulation of these processes leads to chronic inflammation or tissue fibrosis. Wnt ligands are present both in homeostatic and pathological conditions. However, their roles and mechanisms regulating inflammation and tissue repair are being investigated. Here we aim to provide an overview of overarching themes regarding Wnt and macrophages by reviewing the previous literature. We aim to gain future insights into how tissue inflammation, repair, regeneration, and fibrosis events are regulated by macrophages.
Collapse
|
10
|
Aryana IGPS, Rini SS, Soejono CH. The Importance of on Sclerostin as Bone-Muscle Mediator Crosstalk. Ann Geriatr Med Res 2022; 26:72-82. [PMID: 35599457 PMCID: PMC9271392 DOI: 10.4235/agmr.22.0036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/14/2022] [Indexed: 11/12/2022] Open
Abstract
Loss of bone and muscle mass is a frequent aging condition and has become a growing public health problem. The term “osteosarcopenia” denotes close links between bone and muscle. Mechanical exercise was once thought to be the only mechanism of crosstalk between muscle and bone. Sclerostin is an important player in the process of unloading-induced bone loss and plays an important role in mechanotransduction in the bone. Furthermore, bones and muscles are categorized as endocrine organs because they produce hormone-like substances, resulting in “bone-muscle crosstalk.” Sclerostin, an inhibitor of bone development, has recently been shown to play a role in myogenesis. This review discusses the importance of sclerostin in bone-muscle crosstalk.
Collapse
Affiliation(s)
- I Gusti Putu Suka Aryana
- Division of Geriatric Medicine, Department of Internal Medicine, Sanglah Hospital–Faculty of Medicine Udayana University, Bali, Indonesia
- Corresponding Author: I Gusti Putu Suka Aryana, MD, PhD Division of Geriatrics, Department of Internal Medicine, Sanglah Hospital–Faculty of Medicine Udayana University, Jl. Pulau Tarakan No.1, Denpasar 80114, Bali, Indonesia E-mail:
| | - Sandra Surya Rini
- Department of Internal Medicine, North Lombok Regional Hospital, West Nusa Tenggara, Indonesia
| | - Czeresna Heriawan Soejono
- Division of Geriatric Medicine, Department of Internal Medicine, Cipto Mangunkusumo Hospital–Faculty of Medicine University of Indonesia, Jakarta, Indonesia
| |
Collapse
|
11
|
(-) - Epicatechin improves Tibialis anterior muscle repair in CD1 mice with BaCl2-induced damage. J Nutr Biochem 2022; 107:109069. [DOI: 10.1016/j.jnutbio.2022.109069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 02/05/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022]
|
12
|
McKee CM, Chapski DJ, Wehling-Henricks M, Rosa-Garrido M, Kuro-O M, Vondriska TM, Tidball JG. The anti-aging protein Klotho affects early postnatal myogenesis by downregulating Jmjd3 and the canonical Wnt pathway. FASEB J 2022; 36:e22192. [PMID: 35174906 PMCID: PMC9007106 DOI: 10.1096/fj.202101298r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/15/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022]
Abstract
Modulating the number of muscle stems cells, called satellite cells, during early postnatal development produces long-term effects on muscle growth. We tested the hypothesis that high expression levels of the anti-aging protein Klotho in early postnatal myogenesis increase satellite cell numbers by influencing the epigenetic regulation of genes that regulate myogenesis. Our findings show that elevated klotho expression caused a transient increase in satellite cell numbers and slowed muscle fiber growth, followed by a period of accelerated muscle growth that leads to larger fibers. Klotho also transcriptionally downregulated the H3K27 demethylase Jmjd3, leading to increased H3K27 methylation and decreased expression of genes in the canonical Wnt pathway, which was associated with a delay in muscle differentiation. In addition, Klotho stimulation and Jmjd3 downregulation produced similar but not additive reductions in the expression of Wnt4, Wnt9a, and Wnt10a in myogenic cells, indicating that inhibition occurred through a common pathway. Together, our results identify a novel pathway through which Klotho influences myogenesis by reducing the expression of Jmjd3, leading to reductions in the expression of Wnt genes and inhibition of canonical Wnt signaling.
Collapse
Affiliation(s)
- Cynthia M McKee
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
| | - Douglas J Chapski
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, USA
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Thomas M Vondriska
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Departments of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| |
Collapse
|
13
|
Cho HJ, Kim H, Lee YS, Moon SA, Kim JM, Kim H, Kim MJ, Yu J, Kim K, Baek IJ, Lee SH, Ahn KH, Kim S, Kang JS, Koh JM. SLIT3 promotes myogenic differentiation as a novel therapeutic factor against muscle loss. J Cachexia Sarcopenia Muscle 2021; 12:1724-1740. [PMID: 34423586 PMCID: PMC8718016 DOI: 10.1002/jcsm.12769] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/25/2021] [Accepted: 07/10/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Sarcopenia and osteoporosis frequently co-occur in the elderly and have common pathophysiological determinants. Slit guidance ligand 3 (SLIT3) has been recently discovered as a novel therapeutic factor against osteoporosis, and a SLIT3 fragment containing the second leucine-rich repeat domain (LRRD2) had a therapeutic efficacy against osteoporosis. However, a role of SLIT3 in the skeletal muscle is unknown. METHODS Skeletal muscle mass, strength, and/or physical activity were evaluated in Slit3-/- , ovariectomized, and aged mice, based on the measurements of muscle weight and grip strength, Kondziella's inverted hanging test, and/or wheel-running test. Skeletal muscles were also histologically evaluated by haematoxylin and eosin staining and/or immunofluorescence. The ovariectomized and aged mice were intravenously injected with recombinant SLIT3 LRRD2 for 4 weeks. C2C12 cells were used to know cellular effects of SLIT3, such as in vitro myogenesis, fusion, cell viability, and proliferation, and also used to evaluate its molecular mechanisms by immunocytochemistry, immunoprecipitation, western blotting, real-time PCR, siRNA transfection, and receptor-ligand binding ELISA. RESULTS Slit3-deficient mice exhibited decreased skeletal muscle mass, muscle strength, and physical activity. The relative masses of gastrocnemius and soleus were lower in the Slit3-/- mice (0.580 ± 0.039% and 0.033 ± 0.003%, respectively) than those in the WT littermates (0.622 ± 0.043% and 0.038 ± 0.003%, respectively) (all, P < 0.05). Gastrocnemius of Slit3-/- mice showed the reduced number of Type I and Type IIa fibres (all, P < 0.05), but not of Type IIb and Type IIx fibres. SLIT3 activated β-catenin signalling by promoting its release from M-cadherin, thereby increasing myogenin expression to stimulate myoblast differentiation. In vitro experiments involving ROBO2 expression, knockdown, and interaction with SLIT3 indicated that ROBO2 functions as a SLIT3 receptor to aid myoblast differentiation. SLIT3 LRRD2 dissociated M-cadherin-bound β-catenin and up-regulated myogenin expression to increase myoblast differentiation, in a manner similar to full-length SLIT3. Systemic treatment with SLIT3 LRRD2 increased skeletal muscle mass in both ovariectomized and aged mice (all, P < 0.05). The relative masses of gastrocnemius and soleus were higher in the treated aged mice (0.548 ± 0.045% and 0.033 ± 0.005%, respectively) than in the untreated aged mice (0.508 ± 0.016% and 0.028 ± 0.003%, respectively) (all, P < 0.05). SLIT3 LRRD2 treatment increased the hanging duration of the aged mice by approximately 1.7-fold (P < 0.05). CONCLUSIONS SLIT3 plays a sarcoprotective role by activating β-catenin signalling. SLIT3 LRRD2 can potentially be used as a therapeutic agent against muscle loss.
Collapse
Affiliation(s)
- Han Jin Cho
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Hyeonmok Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Young-Sun Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Sung Ah Moon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Jin-Man Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Hanjun Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Min Ji Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea.,Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, South Korea
| | - In-Jeoung Baek
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Sungsub Kim
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
14
|
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: 0.7] [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
|
15
|
Kimura A, Toyoda T, Iwasaki M, Hirama R, Osafune K. Combined Omics Approaches Reveal the Roles of Non-canonical WNT7B Signaling and YY1 in the Proliferation of Human Pancreatic Progenitor Cells. Cell Chem Biol 2020; 27:1561-1572.e7. [PMID: 33125912 DOI: 10.1016/j.chembiol.2020.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023]
Abstract
The proliferation of human pancreatic progenitor cells (PPCs) is critical for developing cell therapies for diabetes. Here, using transcriptome analysis combined with small interfering RNA (siRNA) screening, we revealed that WNT7B is a downstream growth factor of AT7867, a compound known to promote the proliferation of PPCs generated from human pluripotent stem cells. Feeder cell lines stably expressing mouse Wnt7a or Wnt7b, but not other Wnts, enhanced PPC proliferation in the absence of AT7867. Importantly, Wnt7a/b ligands did not activate the canonical Wnt pathway, and PPC proliferation depended on the non-canonical Wnt/PKC pathway. A comparison of the phosphoproteome in response to AT7867 or a newly synthesized AT7867 derivative uncovered the function of YY1 as a transcriptional regulator of WNT7B. Overall, our data highlight unknown roles of non-canonical WNT7B/PKC signaling and YY1 in human PPC proliferation and will contribute to the stable supply of a cell source for pancreatic disease modeling and therapeutic applications.
Collapse
Affiliation(s)
- Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Mio Iwasaki
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Ryusuke Hirama
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kawasaki, Kanagawa 210-8681, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
16
|
Bai Y, Nie H, Wang Z, Yan X. Genome-wide identification and transcriptome-based expression profiling of Wnt gene family in Ruditapes philippinarum. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2020; 35:100709. [PMID: 32688272 DOI: 10.1016/j.cbd.2020.100709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
The Wnt genes encode a set of conserved glycoproteins regulating early development, cell proliferation and differentiation, and tissue regeneration in metazoans. In some mollusks, the knowledge of Wnt gene family has been limited because of the short of the genomic and transcriptomic resources. Ruditapes philippinarum is an economically important bivalve with a variety of shell coloration patterns and ability to regenerate its siphon. To gain a greater understanding of the evolutionary dynamics of Wnt gene family, we carried out a genome-wide identification and phylogenetic analysis of Wnt gene family in R. philippinarum and other four mollusks. A total of 12 Wnt genes were identified in the genome of R. philippinarum, and the dynamic patterns of gene conservation, loss and duplication of Wnt genes were analyzed in mollusks and model organisms. Furthermore, the transcriptome analyses demonstrated the expression profiles of the Wnt genes at different developmental stage, in adult tissues, during siphon regeneration, in four different shell color strains, and at uncolored and colored developmental stages in two different shell color strains. These findings suggest that the expansion of Wnt genes may play vital roles in the larval development, the formation of shell color pattern and siphon regeneration in R. philippinarum. This study provides a valuable insight into Wnt function and evolution in mollusks.
Collapse
Affiliation(s)
- Yitian Bai
- College of Fisheries and Life Science, Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Hongtao Nie
- College of Fisheries and Life Science, Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, Dalian 116023, China.
| | - Zhengxing Wang
- College of Fisheries and Life Science, Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| | - Xiwu Yan
- College of Fisheries and Life Science, Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
17
|
Chen YC, Aui SP, Lai YS, Chang KT. Adult Stem Cells in Hibernation: Future Perspectives of Space Travel. Int J Stem Cells 2019; 12:381-387. [PMID: 31474026 PMCID: PMC6881040 DOI: 10.15283/ijsc19048] [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] [Received: 04/08/2019] [Revised: 06/20/2019] [Accepted: 07/05/2019] [Indexed: 11/09/2022] Open
Abstract
Space traveling is imperative for mankind in the future. Expectedly, hibernation will become an option for space traveler to overcome the endless voyage. With regard to some of the studies pointed out that during hibernation, muscle will undergo atrophy and meantime neurogenesis will reduce, these obstacles were frequently related with stem cell regeneration. Thus, investigation on whether hibernation will lead to dysfunction of stem cell becomes an important issue. By going through four main systems in this article, such as, hematopoietic system, skeletal muscle system, central nervous system and orthopedic system, we are expecting that stem cells regeneration capacity will be affected by hibernation. To date, these researches are majorly the read-out from short term or seasonal hibernating mammals. Proposing and creating a simulated long-term hibernation animal model is turning essential for the further investigation on the effect of longer period of hibernation to human stem cells.
Collapse
Affiliation(s)
- Yu-Chih Chen
- Graduate Institute of Bioresources, National Pingtung University of Science and Technology, Neipu, Taiwan
| | - Shin-Peir Aui
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Neipu, Taiwan
| | - Yin-Siew Lai
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Neipu, Taiwan.,Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, Neipu, Taiwan.,Research Center for Animal Biologics, National Pingtung University of Science and Technology, Neipu, Taiwan
| | - Ko-Tung Chang
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Neipu, Taiwan.,Flow Cytometry Center, Precision Instruments Center, National Pingtung University of Science and Technology, Neipu, Taiwan.,Research Center for Animal Biologics, National Pingtung University of Science and Technology, Neipu, Taiwan
| |
Collapse
|
18
|
Chijimatsu R, Saito T. Mechanisms of synovial joint and articular cartilage development. Cell Mol Life Sci 2019; 76:3939-3952. [PMID: 31201464 PMCID: PMC11105481 DOI: 10.1007/s00018-019-03191-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/30/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022]
Abstract
Articular cartilage is formed at the end of epiphyses in the synovial joint cavity and permanently contributes to the smooth movement of synovial joints. Most skeletal elements develop from transient cartilage by a biological process known as endochondral ossification. Accumulating evidence indicates that articular and growth plate cartilage are derived from different cell sources and that different molecules and signaling pathways regulate these two kinds of cartilage. As the first sign of joint development, the interzone emerges at the presumptive joint site within a pre-cartilage tissue. After that, joint cavitation occurs in the center of the interzone, and the cells in the interzone and its surroundings gradually form articular cartilage and the synovial joint. During joint development, the interzone cells continuously migrate out to the epiphyseal cartilage and the surrounding cells influx into the joint region. These complicated phenomena are regulated by various molecules and signaling pathways, including GDF5, Wnt, IHH, PTHrP, BMP, TGF-β, and FGF. Here, we summarize current literature and discuss the molecular mechanisms underlying joint formation and articular development.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Bone and Cartilage Regenerative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
19
|
Li S, Liu D, Fu Y, Zhang C, Tong H, Li S, Yan Y. Podocan Promotes Differentiation of Bovine Skeletal Muscle Satellite Cells by Regulating the Wnt4-β-Catenin Signaling Pathway. Front Physiol 2019; 10:1010. [PMID: 31447699 PMCID: PMC6692459 DOI: 10.3389/fphys.2019.01010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/22/2019] [Indexed: 02/03/2023] Open
Abstract
Background Small leucine-rich repeat proteins (SLRPs) are highly effective and selective modulators of cell proliferation and differentiation. Podocan is a newly discovered member of the SLRP family. Its potential roles in the differentiation of bovine muscle-derived satellite cells (MDSCs) and its underlying functional mechanism remain unclear. Our study aimed to characterize the function of the podocan gene in the differentiation of bovine MDSCs and to clarify the molecular mechanism by which podocan functions in order to contribute to a better understanding of the molecular mechanism by which extracellular matrix promotes bovine MDSC differentiation and provide a theoretical basis for the improvement of beef quality. Methods Bovine MDSCs were transfected with vectors to overexpress or inhibit podocan, and podocan protein was added to differentiation culture medium. qRT-PCR, western blotting, and immunofluorescence were performed to investigate the effects of podocan on MDSC differentiation. Confocal microscopy and western blotting were used to assess the nuclear translocation and expression of β-catenin. An inhibitor and activator of β-catenin were used to assess the effects of the Wnt/β-catenin signaling pathway on MDSC differentiation. We inhibited β-catenin while overexpressing podocan in MDSCs. Then, we performed mass spectrometry to identify which proteins interact with podocan to regulate the Wnt/β-catenin signaling pathway. Finally, we confirmed the relationship between podocan and Wnt4 by co-immunoprecipitation and western blotting. Results Podocan protein expression increased significantly during bovine MDSC differentiation. Differentiation of bovine MDSC was promoted and suppressed by podocan overexpression or inhibition, respectively. Podocan was also shown to modulate the Wnt/β-catenin signaling pathway. Treatment of bovine MDSCs with β-catenin inhibitor and activator showed that the Wnt/β-catenin pathway is involved in bovine MDSC differentiation. Furthermore, the effect of podocan on bovine MDSC differentiation was suppressed when this pathway was inhibited. We also found that podocan interacts with Wnt4. When Wnt4 was inhibited, podocan-induced promotion of bovine MDSC differentiation was attenuated through Wnt/β-catenin signaling. Conclusion Podocan regulates Wnt/β-catenin through Wnt4 to promote bovine MDSC differentiation.
Collapse
Affiliation(s)
- Shuang Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| | - Dan Liu
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| | - Yuying Fu
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| | - Chunyu Zhang
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| | - Huili Tong
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| | - Shufeng Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| | - Yunqin Yan
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, China
| |
Collapse
|
20
|
Hung LY, Johnson JL, Ji Y, Christian DA, Herbine KR, Pastore CF, Herbert DR. Cell-Intrinsic Wnt4 Influences Conventional Dendritic Cell Fate Determination to Suppress Type 2 Immunity. THE JOURNAL OF IMMUNOLOGY 2019; 203:511-519. [PMID: 31175162 DOI: 10.4049/jimmunol.1900363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/15/2019] [Indexed: 01/26/2023]
Abstract
Whether conventional dendritic cells (cDC) acquire subset identity under direction of Wnt family glycoproteins is unknown. We demonstrate that Wnt4, a β-catenin-independent Wnt ligand, is produced by both hematopoietic and nonhematopoietic cells and is both necessary and sufficient for preconventional DC1/cDC1 maintenance. Whereas bone marrow cDC precursors undergo phosphoJNK/c-Jun activation upon Wnt4 treatment, loss of cDC Wnt4 in CD11cCreWnt4flox/flox mice impaired differentiation of CD24+, Clec9A+, CD103+ cDC1 compared with CD11cCre controls. Conversely, single-cell RNA sequencing analysis of bone marrow revealed a 2-fold increase in cDC2 gene signature genes, and flow cytometry demonstrated increased numbers of SIRP-α+ cDC2 amid lack of Wnt4. Increased cDC2 numbers due to CD11c-restricted Wnt4 deficiency increased IL-5 production, group 2 innate lymphoid cell expansion, and host resistance to the hookworm parasite Nippostrongylus brasiliensis Collectively, these data uncover a novel and unexpected role for Wnt4 in cDC subset differentiation and type 2 immunity.
Collapse
Affiliation(s)
- Li-Yin Hung
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - John L Johnson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Yingbiao Ji
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Karl R Herbine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Christopher F Pastore
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - De'Broski R Herbert
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
21
|
Cui S, Li L, Yu RT, Downes M, Evans RM, Hulin JA, Makarenkova HP, Meech R. β-Catenin is essential for differentiation of primary myoblasts via cooperation with MyoD and α-catenin. Development 2019; 146:dev.167080. [PMID: 30683662 DOI: 10.1242/dev.167080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022]
Abstract
Canonical Wnts promote myoblast differentiation; however, the role of β-catenin in adult myogenesis has been contentious, and its mechanism(s) unclear. Using CRISPR-generated β-catenin-null primary adult mouse myoblasts, we found that β-catenin was essential for morphological differentiation and timely deployment of the myogenic gene program. Alignment, elongation and fusion were grossly impaired in null cells, and myogenic gene expression was not coordinated with cytoskeletal and membrane remodeling events. Rescue studies and genome-wide analyses extended previous findings that a β-catenin-TCF/LEF interaction is not required for differentiation, and that β-catenin enhances MyoD binding to myogenic loci. We mapped cellular pathways controlled by β-catenin and defined novel targets in myoblasts, including the fusogenic genes myomaker and myomixer. We also showed that interaction of β-catenin with α-catenin was important for efficient differentiation. Overall the study suggests dual roles for β-catenin: a TCF/LEF-independent nuclear function that coordinates an extensive network of myogenic genes in cooperation with MyoD; and an α-catenin-dependent membrane function that helps control cell-cell interactions. β-Catenin-TCF/LEF complexes may function primarily in feedback regulation to control levels of β-catenin and thus prevent precocious/excessive myoblast fusion.
Collapse
Affiliation(s)
- Shuang Cui
- Department of Clinical Pharmacology, Flinders University, Bedford Park, SA 5042, Australia.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Liang Li
- Department of Biochemistry, Flinders University, Bedford Park, SA 5042 and Department of Biochemistry, University of Adelaide, North Tce, Adelaide, SA 5005, Australia
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute, La Jolla, CA 92037, USA.,Howard Hughes Medical Institute, Salk Institute, La Jolla, CA 92037, USA
| | - Julie-Ann Hulin
- Department of Clinical Pharmacology, Flinders University, Bedford Park, SA 5042, Australia
| | - Helen P Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Robyn Meech
- Department of Clinical Pharmacology, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
22
|
Tsukamoto M, Wang KY, Tasaki T, Murata Y, Okada Y, Yamanaka Y, Nakamura E, Yamada S, Izumi H, Zhou Q, Azuma K, Sasaguri Y, Kohno K, Sakai A. Findings as a starting point to unravel the underlying mechanisms of in vivo interactions involving Wnt10a in bone, fat and muscle. Bone 2019; 120:75-84. [PMID: 30315998 DOI: 10.1016/j.bone.2018.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/08/2018] [Accepted: 10/08/2018] [Indexed: 12/15/2022]
Abstract
Wnt10a is a member of the WNT family. Although deficiency of this gene causes symptoms related to teeth, hair, nails, and skin, we recently demonstrated a new phenotype of Wnt10a knockout (KO) mice involving bone and fat. The in vivo effect of the Wnt10a gene on bone and fat is unclear, and the relationship between bone/fat and muscle in Wnt10a signaling is also interesting. We aimed to evaluate the tissue changes in Wnt10a KO mice compared to wild-type mice and show the findings as a starting point to unravel the underlying mechanisms of in vivo interactions involving Wnt10a in bone, fat and muscle. Trabecular bone loss in the lower limbs of Wnt10a mice and decreased bone mineralization were observed. The adipose tissue in bone marrow was also decreased, and adipocyte differentiation was reduced. The body fat mass in Wnt10a KO mice was decreased, and white adipocytes in subcutaneous fat were converted to beige adipocytes. The muscle weight of the lower limbs was not decreased despite trabecular bone loss, but Gdf8/myostatin expression was reduced in the subcutaneous fat and gastrocnemius muscles of Wnt10a KO mice. Thus, in vivo deletion of Wnt10a inhibited osteogenic activity, promoted beige adipogenesis of white adipocytes and maintained muscle mass. These results suggest that regulation of Gdf8 by Wnt10a may help maintain the muscle mass of Wnt10a KO mice. This study was the first to histologically evaluate the bone, fat and muscle phenotypes of Wnt10a KO mice. The results of this study, which were obtained by investigating the three tissues together, could influence the understanding of in vivo interactions involving the Wnt10a gene.
Collapse
Affiliation(s)
- Manabu Tsukamoto
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health University, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Ke-Yong Wang
- Shared-Use Research Center, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | - Takashi Tasaki
- Department of Pathology and Cell Biology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yoichi Murata
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health University, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yasuaki Okada
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health University, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yoshiaki Yamanaka
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health University, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Eiichiro Nakamura
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health University, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Sohsuke Yamada
- Department of Pathology and Laboratory Medicine, Kanazawa Medical University, 1-1 Uchinada, Ishikawa 920-0293, Japan
| | - Hiroto Izumi
- Department of Occupational Pneumology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Qian Zhou
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Kagaku Azuma
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | - Yasuyuki Sasaguri
- Department of Pathology and Cell Biology, School of Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan; Laboratory of Pathology, Fukuoka Tokushukai Hospital, Fukuoka 816-0864, Japan
| | | | - Akinori Sakai
- Department of Orthopaedic Surgery, School of Medicine, University of Occupational and Environmental Health University, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| |
Collapse
|
23
|
Chang CN, Kioussi C. Location, Location, Location: Signals in Muscle Specification. J Dev Biol 2018; 6:E11. [PMID: 29783715 PMCID: PMC6027348 DOI: 10.3390/jdb6020011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
Muscles control body movement and locomotion, posture and body position and soft tissue support. Mesoderm derived cells gives rise to 700 unique muscles in humans as a result of well-orchestrated signaling and transcriptional networks in specific time and space. Although the anatomical structure of skeletal muscles is similar, their functions and locations are specialized. This is the result of specific signaling as the embryo grows and cells migrate to form different structures and organs. As cells progress to their next state, they suppress current sequence specific transcription factors (SSTF) and construct new networks to establish new myogenic features. In this review, we provide an overview of signaling pathways and gene regulatory networks during formation of the craniofacial, cardiac, vascular, trunk, and limb skeletal muscles.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
- Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
24
|
Hitachi K, Tsuchida K. Myostatin-deficiency in mice increases global gene expression at the Dlk1-Dio3 locus in the skeletal muscle. Oncotarget 2018; 8:5943-5953. [PMID: 27992376 PMCID: PMC5351603 DOI: 10.18632/oncotarget.13966] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/08/2016] [Indexed: 12/31/2022] Open
Abstract
Myostatin, a member of the transforming growth factor-beta superfamily, is a negative regulator of skeletal muscle growth and development. Myostatin inhibition leads to increased skeletal muscle mass in mammals; hence, myostatin is considered a potential therapeutic target for skeletal muscle wasting. However, downstream molecules of myostatin in the skeletal muscle have not been fully elucidated. Here, we identified the Dlk1-Dio3 locus at the mouse chromosome 12qF1, also called as the callipyge locus in sheep, as a novel downstream target of myostatin. In skeletal muscle of myostatin knockout mice, the expression of mature miRNAs at the Dlk1-Dio3 locus was significantly increased. The increased miRNA levels are caused by the transcriptional activation of the Dlk1-Dio3 locus, because a significant increase in the primary miRNA transcript was observed in myostatin knockout mice. In addition, we found increased expression of coding and non-coding genes (Dlk1, Gtl2, Rtl1/Rtl1as, and Rian) at the Dlk1-Dio3 locus in myostatin-deficient skeletal muscle. Moreover, epigenetic changes, associated with the regulation of the Dlk1-Dio3 locus, were observed in myostatin knockout mice. Taken together, this is the first report demonstrating the role of myostatin in regulating the Dlk1-Dio3 (the callipyge) locus in the skeletal muscle.
Collapse
Affiliation(s)
- Keisuke Hitachi
- Division for Therapies Against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kunihiro Tsuchida
- Division for Therapies Against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
25
|
Girardi F, Le Grand F. Wnt Signaling in Skeletal Muscle Development and Regeneration. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:157-179. [DOI: 10.1016/bs.pmbts.2017.11.026] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
26
|
Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:153-206. [DOI: 10.1007/978-981-13-1435-3_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Dasarathy S. Myostatin and beyond in cirrhosis: all roads lead to sarcopenia. J Cachexia Sarcopenia Muscle 2017; 8:864-869. [PMID: 29168629 PMCID: PMC5700432 DOI: 10.1002/jcsm.12262] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Srinivasan Dasarathy
- Professor of Medicine, Cleveland Clinic Lerner College of Medicine; Director, Liver Metabolism Research; Staff, Departments of Gastroenterology, Hepatology and Pathobiology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
28
|
Cox B, Roose H, Vennekens A, Vankelecom H. Pituitary stem cell regulation: who is pulling the strings? J Endocrinol 2017; 234:R135-R158. [PMID: 28615294 DOI: 10.1530/joe-17-0083] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/14/2017] [Indexed: 12/28/2022]
Abstract
The pituitary gland plays a pivotal role in the endocrine system, steering fundamental processes of growth, metabolism, reproduction and coping with stress. The adult pituitary contains resident stem cells, which are highly quiescent in homeostatic conditions. However, the cells show marked signs of activation during processes of increased cell remodeling in the gland, including maturation at neonatal age, adaptation to physiological demands, regeneration upon injury and growth of local tumors. Although functions of pituitary stem cells are slowly but gradually uncovered, their regulation largely remains virgin territory. Since postnatal stem cells in general reiterate embryonic developmental pathways, attention is first being given to regulatory networks involved in pituitary embryogenesis. Here, we give an overview of the current knowledge on the NOTCH, WNT, epithelial-mesenchymal transition, SHH and Hippo pathways in the pituitary stem/progenitor cell compartment during various (activation) conditions from embryonic over neonatal to adult age. Most information comes from expression analyses of molecular components belonging to these networks, whereas functional extrapolation is still very limited. From this overview, it emerges that the 'big five' embryonic pathways are indeed reiterated in the stem cells of the 'lazy' homeostatic postnatal pituitary, further magnified en route to activation in more energetic, physiological and pathological remodeling conditions. Increasing the knowledge on the molecular players that pull the regulatory strings of the pituitary stem cells will not only provide further fundamental insight in postnatal pituitary homeostasis and activation, but also clues toward the development of regenerative ideas for improving treatment of pituitary deficiency and tumors.
Collapse
Affiliation(s)
- Benoit Cox
- Department of Development and RegenerationCluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven, Belgium
| | - Heleen Roose
- Department of Development and RegenerationCluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven, Belgium
| | - Annelies Vennekens
- Department of Development and RegenerationCluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven, Belgium
| | - Hugo Vankelecom
- Department of Development and RegenerationCluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
29
|
Increased Myogenic and Protein Turnover Signaling in Skeletal Muscle of Chronic Obstructive Pulmonary Disease Patients With Sarcopenia. J Am Med Dir Assoc 2017; 18:637.e1-637.e11. [PMID: 28578881 DOI: 10.1016/j.jamda.2017.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 04/13/2017] [Accepted: 04/17/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Sarcopenia was recently recognized as an independent condition by an International Classification of Diseases, Tenth Revision, Clinical Modification code, and is a frequently observed comorbidity in chronic obstructive pulmonary disease (COPD). Muscle mass is primarily dictated by the balance between protein degradation and synthesis, but their relative contribution to sarcopenia is unclear. OBJECTIVE We aimed to assess potential differential molecular regulation of protein degradation and synthesis, as well as myogenesis, in the skeletal muscle of COPD patients with and without sarcopenia. METHODS Muscle biopsies were obtained from the vastus lateralis muscle. Patients with COPD were clustered based on sarcopenia defined by low appendicular skeletal muscle mass index (nonsarcopenic COPD, n = 53; sarcopenic COPD, n = 39), and compared with healthy nonsarcopenic controls (n = 13). The mRNA and protein expression of regulators and mediators of ubiquitin-proteasome system (UPS), autophagy-lysosome system (autophagy), and protein synthesis were analyzed. Furthermore, mRNA expression of myogenesis markers was assessed. RESULTS UPS signaling was unaltered, whereas indices of UPS regulation (eg, FOXO1 protein; p-FOXO3/FOXO3), autophagy signaling (eg, LC3BII/I; p-ULK1[Ser757]/ULK1), and protein synthesis signaling (eg, AKT1; p-GSK3B/GSK3B; p-4E-BP1/4E-BP1) were increased in COPD. These alterations were even more pronounced in COPD patients with sarcopenia (eg, FOXO1 protein; p-FOXO1/FOXO1; LC3BII/I; p-ULK(Ser555); p-AKT1/AKT1; AKT1; p-4E-BP1). Furthermore, myogenic signaling (eg, MYOG) was increased in COPD despite a concomitant increase of myostatin (MSTN) mRNA expression, with no difference between sarcopenic and nonsarcopenic COPD patients. CONCLUSION Together with elevated myogenic signaling, the increase in muscle protein turnover signaling in COPD, which is even more prominent in COPD patients with sarcopenia, reflects molecular alterations associated with muscle repair and remodeling.
Collapse
|
30
|
Liu M, Lee C, Laron D, Zhang N, Waldorff EI, Ryaby JT, Feeley B, Liu X. Role of pulsed electromagnetic fields (PEMF) on tenocytes and myoblasts-potential application for treating rotator cuff tears. J Orthop Res 2017; 35:956-964. [PMID: 27138553 DOI: 10.1002/jor.23278] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/25/2016] [Indexed: 02/04/2023]
Abstract
The post-surgery integrity of the tendons and muscle quality are the two major factors in success of rotator cuff (RC) repair. Though surgical techniques for rotator cuff repair have significantly improved in the past two decades, there are no effective treatments to improve tendon-to-bone healing and muscle quality after repair at this point in time. Pulsed electromagnetic fields (PEMF) have previously been used for promoting fracture healing. Previous studies have shown that PEMF has a positive role in promoting osteoblast precursors proliferation and differentiation. However, PEMFs effect on tenocytes and muscle cells has not been determined fully yet. The purpose of this study is to define the role of a commercially available PEMF on tenocytes and myoblasts growth and differentiation in vitro. Human rotator cuff tenocytes and C2C12 murine myoblasts were cultured and treated with PEMF for 2 weeks under regular and inflammatory conditions. Our results showed that 2 weeks treatment of PEMF enhanced gene expressions of growth factors in human rotator cuff tenocytes under inflammatory conditions. PEMF significantly enhanced C2C12 myotube formation under normal and inflammatory conditions. Results from this study suggest that PEMF has a positive role in promoting tenocyte gene expression and myoblast differentiation. Therefore, PEMF may potentially serve as a non-operative treatment to improve clinical incomes rotator cuff tendon repairs. Results © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:956-964, 2017.
Collapse
Affiliation(s)
- Mengyao Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, 1700 Owens Street, Room 364, San Francisco, California, 94153.,Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Carlin Lee
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, 1700 Owens Street, Room 364, San Francisco, California, 94153.,Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Dominique Laron
- Department of Orthopaedic Surgery, University of California, San Francisco, California
| | | | | | | | - Brian Feeley
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, 1700 Owens Street, Room 364, San Francisco, California, 94153.,Department of Orthopaedic Surgery, University of California, San Francisco, California
| | - Xuhui Liu
- Department of Veterans Affairs, San Francisco Veterans Affairs Medical Center, 1700 Owens Street, Room 364, San Francisco, California, 94153.,Department of Orthopaedic Surgery, University of California, San Francisco, California
| |
Collapse
|
31
|
Huraskin D, Eiber N, Reichel M, Zidek LM, Kravic B, Bernkopf D, von Maltzahn J, Behrens J, Hashemolhosseini S. Wnt/β-catenin signaling via Axin2 is required for myogenesis and, together with YAP/Taz and Tead1, active in IIa/IIx muscle fibers. Development 2016; 143:3128-42. [DOI: 10.1242/dev.139907] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 07/13/2016] [Indexed: 12/18/2022]
Abstract
Canonical Wnt/β-catenin signaling plays an important role in myogenic differentiation, but its physiological role in muscle fibers remains elusive. Here, we studied activation of Wnt/β-catenin signaling in adult muscle fibers and muscle stem cells in an Axin2 reporter mouse. Axin2 is a negative regulator and a target of Wnt/β-catenin signaling. In adult muscle fibers, Wnt/β-catenin signaling is only detectable in a subset of fast fibers that have a significantly smaller diameter than other fast fibers. In the same fibers, immunofluorescence staining for YAP/Taz and Tead1 was detected. Wnt/β-catenin signaling was absent in quiescent and activated satellite cells. Upon injury, Wnt/β-catenin signaling was detected in muscle fibers with centrally located nuclei. During differentiation of myoblasts expression of Axin2, but not of Axin1, increased together with Tead1 target gene expression. Furthermore, absence of Axin1 and Axin2 interfered with myoblast proliferation and myotube formation, respectively. Treatment with the canonical Wnt3a ligand also inhibited myotube formation. Wnt3a activated TOPflash and Tead1 reporter activity, whereas neither reporter was activated in the presence of Dkk1, an inhibitor of canonical Wnt signaling. We propose that Axin2-dependent Wnt/β-catenin signaling is involved in myotube formation and, together with YAP/Taz/Tead1, associated with reduced muscle fiber diameter of a subset of fast fibers.
Collapse
Affiliation(s)
- Danyil Huraskin
- Institute of Biochemistry, Fahrstrasse 17, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Nane Eiber
- Institute of Biochemistry, Fahrstrasse 17, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Martin Reichel
- Nikolaus-Fiebiger-Center of Molecular Medicine, Glückstrasse 6, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Laura M. Zidek
- Leibniz Institute for Age Research/Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, Jena D-07745, Germany
| | - Bojana Kravic
- Institute of Biochemistry, Fahrstrasse 17, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Dominic Bernkopf
- Nikolaus-Fiebiger-Center of Molecular Medicine, Glückstrasse 6, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Julia von Maltzahn
- Leibniz Institute for Age Research/Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, Jena D-07745, Germany
| | - Jürgen Behrens
- Nikolaus-Fiebiger-Center of Molecular Medicine, Glückstrasse 6, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| | - Said Hashemolhosseini
- Institute of Biochemistry, Fahrstrasse 17, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen D-91054, Germany
| |
Collapse
|
32
|
Mammary Development and Breast Cancer: A Wnt Perspective. Cancers (Basel) 2016; 8:cancers8070065. [PMID: 27420097 PMCID: PMC4963807 DOI: 10.3390/cancers8070065] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 06/30/2016] [Accepted: 07/07/2016] [Indexed: 12/21/2022] Open
Abstract
The Wnt pathway has emerged as a key signaling cascade participating in mammary organogenesis and breast oncogenesis. In this review, we will summarize the current knowledge of how the pathway regulates stem cells and normal development of the mammary gland, and discuss how its various components contribute to breast carcinoma pathology.
Collapse
|
33
|
Hulin JA, Nguyen TDT, Cui S, Marri S, Yu RT, Downes M, Evans RM, Makarenkova H, Meech R. Barx2 and Pax7 Regulate Axin2 Expression in Myoblasts by Interaction with β-Catenin and Chromatin Remodelling. Stem Cells 2016; 34:2169-82. [PMID: 27144473 PMCID: PMC5019118 DOI: 10.1002/stem.2396] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 02/15/2016] [Accepted: 04/14/2016] [Indexed: 12/13/2022]
Abstract
Satellite cells are the resident stem cells of skeletal muscle; quiescent in adults until activated by injury to generate proliferating myoblasts. The canonical Wnt signalling pathway, mediated by T-cell factor/lymphoid enhancer factor (TCF/LEF) and β-catenin effector proteins, controls myoblast differentiation in vitro, and recent work suggests that timely termination of the Wnt/β-catenin signal is important for normal adult myogenesis. We recently identified the Barx2 and Pax7 homeobox proteins as novel components of the Wnt effector complex. Here, we examine molecular and epigenetic mechanisms by which Barx2 and Pax7 regulate the canonical Wnt target gene Axin2, which mediates critical feedback to terminate the transcriptional response to Wnt signals. Barx2 is recruited to the Axin2 gene via TCF/LEF binding sites, recruits β-catenin and the coactivator GRIP-1, and induces local H3K-acetylation. Barx2 also promotes nuclear localization of β-catenin. Conversely, Pax7 represses Axin2 promoter/intron activity and inhibits Barx2-mediated H3K-acetylation via the corepressor HDAC1. Wnt3a not only induces Barx2 mRNA, but also stabilises Barx2 protein in myoblasts; conversely, Wnt3a potently inhibits Pax7 protein expression. As Barx2 promotes myogenic differentiation and Pax7 suppresses it, this novel posttranscriptional regulation of Barx2 and Pax7 by Wnt3a may be involved in the specification of differentiation-competent and -incompetent myoblast populations. Finally, we propose a model for dual function of Barx2 downstream of Wnt signals: activation of myogenic target genes in association with canonical myogenic regulatory factors, and regulation of the negative feedback loop that limits the response of myoblasts to Wnt signals via direct interaction of Barx2 with the TCF/β-catenin complex. Stem Cells 2016;34:2169-2182.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Department of Clinical Pharmacology, Flinders University, Bedford Park, South Australia, Australia
| | - Thi Diem Tran Nguyen
- Department of Clinical Pharmacology, Flinders University, Bedford Park, South Australia, Australia.,Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - Shuang Cui
- Department of Clinical Pharmacology, Flinders University, Bedford Park, South Australia, Australia
| | - Shashikanth Marri
- Department of Molecular Medicine, Flinders University, Bedford Park, South Australia, Australia
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute, La Jolla, California, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute, La Jolla, California, USA
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute, La Jolla, California, USA.,Howard Hughes Medical Institute, Salk Institute, La Jolla, California, USA
| | - Helen Makarenkova
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Robyn Meech
- Department of Clinical Pharmacology, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
34
|
Coulton G, Hou Y, Mirczuk SM, Allen SP. Raf kinase inhibitor protein1 is a myogenic inhibitor with conserved function in avians and mammals. Dev Dyn 2016; 245:902-12. [PMID: 27240024 DOI: 10.1002/dvdy.24421] [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: 12/16/2015] [Revised: 04/19/2016] [Accepted: 05/09/2016] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Raf Kinase Inhibitor Protein1 (RKIP) is a tumor suppressor that is present in several adult tissues. It functions as an inhibitor of both Raf/Mek/Erk and NFĸB signaling when unphosphorylated, but following phosphorylation the ability to inhibit Raf/Mek/Erk signaling is lost and RKIP becomes an activator of G-protein coupled receptor signaling. In neonates and adults, RKIP is known to be expressed in muscle; however, its physiological function is currently unknown. RESULTS In this study, we show by in situ hybridization and immunofluorescence that RKIP is also expressed in developing chick embryonic muscle, and mouse C2C12 myoblasts. Furthermore, we demonstrate that, in these systems, it functions as an inhibitor of myogenesis: increased levels of RKIP suppress myotube differentiation whereas decreasing RKIP promotes differentiation. Additionally, we show that the ability of RKIP to inhibit myogenesis is dependent upon its phosphorylation state as only the nonphosphorylated form of RKIP suppresses myogenesis. CONCLUSIONS This study, therefore, clearly demonstrates that RKIP has conserved functions as a myogenic inhibitor in both mammalian and avian muscle. Developmental Dynamics 245:902-912, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gary Coulton
- Institute for Infection and Immunity, St Georges University of London, London, United Kingdom
| | - Yanwen Hou
- Institute for Infection and Immunity, St Georges University of London, London, United Kingdom
| | - Samantha M Mirczuk
- Comparative Biomedical Sciences, The Royal Veterinary College, Camden, London, United Kingdom
| | - Steven P Allen
- Comparative Biomedical Sciences, The Royal Veterinary College, Camden, London, United Kingdom
| |
Collapse
|
35
|
β-Catenin Activation in Muscle Progenitor Cells Regulates Tissue Repair. Cell Rep 2016; 15:1277-90. [PMID: 27134174 DOI: 10.1016/j.celrep.2016.04.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 02/08/2016] [Accepted: 03/31/2016] [Indexed: 11/23/2022] Open
Abstract
Skeletal muscle regeneration relies on a pool of resident muscle stem cells called satellite cells (MuSCs). Following injury-induced destruction of the myofibers, quiescent MuSCs are activated and generate transient amplifying progenitors (myoblasts) that will fuse to form new myofibers. Here, we focus on the canonical Wnt signaling pathway and find that either conditional β-catenin disruption or activation in adult MuSCs results in perturbation of muscle regeneration. Using both in vivo and in vitro approaches, we observed that myoblasts lacking β-catenin show delayed differentiation, whereas myoblasts with constitutively active β-catenin undergo precocious growth arrest and differentiation. Transcriptome analysis further demonstrated that Wnt/β-catenin signaling interacts with multiple pathways and, more specifically, TGF-β signaling. Indeed, exogenous TGF-β2 stimulation restores the regenerative potential of muscles with targeted β-catenin disruption in MuSCs. We conclude that a precise level of β-catenin activity is essential for regulating the amplification and differentiation of MuSC descendants during adult myogenesis.
Collapse
|
36
|
A p38 Mitogen-Activated Protein Kinase-Regulated Myocyte Enhancer Factor 2-β-Catenin Interaction Enhances Canonical Wnt Signaling. Mol Cell Biol 2015; 36:330-46. [PMID: 26552705 DOI: 10.1128/mcb.00832-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
Canonical Wnt/β-catenin signaling plays a major role in various biological contexts, such as embryonic development, cell proliferation, and cancer progression. Previously, a connection between p38 mitogen-activated protein kinase (MAPK) signaling and Wnt-mediated activation of β-catenin was implied but poorly understood. In the present study, we investigated potential cross talk between p38 MAPK and Wnt/β-catenin signaling. Here we show that a loss of p38 MAPK α/β function reduces β-catenin nuclear accumulation in Wnt3a-stimulated primary vascular smooth muscle cells (VSMCs). Conversely, active p38 MAPK signaling increases β-catenin nuclear localization and target gene activity in multiple cell types. Furthermore, the effect of p38 MAPK α/β on β-catenin activity is mediated through phosphorylation of a key p38 MAPK target, myocyte enhancer factor 2 (MEF2). Here we report a p38 MAPK-mediated, phosphorylation-dependent interaction between MEF2 and β-catenin in multiple cell types and primary VSMCs that results in (i) increased β-catenin nuclear retention, which is reversed by small interfering RNA (siRNA)-mediated MEF2 gene silencing; (ii) increased activation of MEF2 and Wnt/β-catenin target genes; and (iii) increased Wnt-stimulated cell proliferation. These observations provide mechanistic insight into a fundamental level of cross talk between p38 MAPK/MEF2 signaling and canonical Wnt signaling.
Collapse
|
37
|
Abstract
UNLABELLED Skeletal muscle satellite cells (SCs) are involved in muscle growth and repair. However, clarification of their behavior in hibernating mammals is lacking. The aim of this study was to quantify SCs and total myonuclei in hibernator muscle during different phases of the torpor-arousal cycle. Skeletal muscle was collected from thirteen-lined ground squirrels, Ictidomys tridecemlineatus, at five timepoints during hibernation: control euthermic [CON, stable body temperature (Tb)], early torpor (ET, within 24h), late torpor (LT, 5+ consecutive days), early arousal (EA, increased respiratory rate >60 breaths/min, Tb 9-12°C) and interbout arousal (IA, euthermic Tb). Protein levels of p21, Myf5, Wnt4, and β-catenin were determined by western blotting. SCs (Pax7(+)) and myonuclei were identified using immunohistochemistry. Over the torpor-arousal cycle, myonuclei/fiber remained unchanged. However, the percentage of SCs increased significantly during ET (7.35±1.04% vs. CONTROL 4.18±0.58%; p<0.05) and returned to control levels during LT. This coincided with a 224% increase in p21 protein during ET. Protein levels of Wnt4 did not change throughout, whereas Myf5 was lower during EA (p<0.08) and IA (p<0.05). Compared to torpor, β-catenin increased by 247% and 279% during EA and IA, respectively (p<0.05). In conclusion, SCs were not dormant during hibernation and increased numbers of SC during ET corresponded with elevated amounts of p21 suggesting that cell cycle control may explain the SC return to baseline levels during late torpor. Despite relatively low Tb during early arousal, active control of quiescence by Myf5 is reduced.
Collapse
|
38
|
Spillane M, Schwarz N, Willoughby DS. Upper-body resistance exercise augments vastus lateralis androgen receptor-DNA binding and canonical Wnt/β-catenin signaling compared to lower-body resistance exercise in resistance-trained men without an acute increase in serum testosterone. Steroids 2015; 98:63-71. [PMID: 25742735 DOI: 10.1016/j.steroids.2015.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/12/2015] [Accepted: 02/20/2015] [Indexed: 10/23/2022]
Abstract
The purpose of the study was to determine the effect of single bouts of lower-body (LB) and upper- and lower-body (ULB) resistance exercise on serum testosterone concentrations and the effects on muscle testosterone, dihydrotestosterone (DHT), androgen receptor (AR) protein content, and AR-DNA binding. A secondary purpose was to determine the effects on serum wingless-type MMTV integration site (Wnt4) levels and skeletal muscle β-catenin content. In a randomized cross-over design, exercise bouts consisted of a LB and ULB protocol, and each bout was separated by 1 week. Blood and muscle samples were obtained before exercise and 3 and 24h post-exercise; blood samples were also obtained at 0.5, 1, and 2 h post-exercise. Statistical analyses were performed by separate two-way factorial analyses of variance (ANOVA) with repeated measures. No significant differences from baseline were observed in serum total and free testosterone and skeletal muscle testosterone and DHT with either protocol (p>0.05). AR protein was significantly increased at 3 h post-exercise and decreased at 24 h post-exercise for ULB, whereas AR-DNA binding was significantly increased at 3 and 24h post-exercise (p<0.05). In response to ULB, serum Wnt4 was significantly increased at 0.5, 1, and 2 h post-exercise (p<0.05) and β-catenin was significantly increased at 3 and 24 h post-exercise (p<0.05). It was concluded that, despite a lack of increase in serum testosterone and muscle androgen concentrations from either mode of resistance exercise, ULB resistance exercise increased Wnt4/β-catenin signaling and AR-DNA binding.
Collapse
Affiliation(s)
- Mike Spillane
- Department of Health, Physical Education, and Leisure Studies, University of South Alabama, Mobile, AL 36688, USA
| | - Neil Schwarz
- Department of Health, Physical Education, and Leisure Studies, University of South Alabama, Mobile, AL 36688, USA
| | - Darryn S Willoughby
- Exercise and Biochemical Nutrition Lab, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76798, USA.
| |
Collapse
|
39
|
Katase N, Terada K, Suzuki T, Nishimatsu SI, Nohno T. miR-487b, miR-3963 and miR-6412 delay myogenic differentiation in mouse myoblast-derived C2C12 cells. BMC Cell Biol 2015; 16:13. [PMID: 25925429 PMCID: PMC4433089 DOI: 10.1186/s12860-015-0061-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/22/2015] [Indexed: 12/17/2022] Open
Abstract
Background Skeletal muscle differentiation is a multistep, complex pathway in which several important signaling molecules are involved. Recently, microRNAs (miRNAs), endogenous non-coding small RNAs that regulate mRNAs, have been proposed to be involved in skeletal muscle differentiation. In this study, we identified skeletal muscle differentiation-associated miRNAs by comparing miRNA expression profiles between C2C12 cells and Wnt4 over-expressing C2C12 cells (W4-08), which can spontaneously differentiate into myotubes. Results We identified miR-206, miR-133a, and miR-133b as up-regulated miRNAs and miR-487b, miR-3963 and miR-6412 as down-regulated miRNAs in differentiating cells. We focused on the down-regulated miRNAs because their functions were largely unknown. Transfection of mimics of these miRNAs into C2C12 cells resulted in significantly reduced expression of myogenic differentiation markers, including troponin T and myosin heavy chain fast type and slow type, but did not affect the expression of the myogenic transcription factors, MyoD and myogenin. Conclusions These miRNAs were characterized as new myogenic differentiation-associated miRNAs which may delay late myogenic differentiation or maturation. Electronic supplementary material The online version of this article (doi:10.1186/s12860-015-0061-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naoki Katase
- Department of Molecular and Developmental Biology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Kumiko Terada
- Department of Molecular and Developmental Biology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Takahiro Suzuki
- Department of Molecular and Developmental Biology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Shin-ichiro Nishimatsu
- Department of Molecular and Developmental Biology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Tsutomu Nohno
- Department of Molecular and Developmental Biology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| |
Collapse
|
40
|
Zhuang L, Hulin JA, Gromova A, Tran Nguyen TD, Yu RT, Liddle C, Downes M, Evans RM, Makarenkova HP, Meech R. Barx2 and Pax7 have antagonistic functions in regulation of wnt signaling and satellite cell differentiation. Stem Cells 2015; 32:1661-73. [PMID: 24753152 DOI: 10.1002/stem.1674] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 12/16/2013] [Accepted: 01/16/2012] [Indexed: 11/05/2022]
Abstract
The canonical Wnt signaling pathway is critical for myogenesis and can induce muscle progenitors to switch from proliferation to differentiation; how Wnt signals integrate with muscle-specific regulatory factors in this process is poorly understood. We previously demonstrated that the Barx2 homeobox protein promotes differentiation in cooperation with the muscle regulatory factor (MRF) MyoD. Pax7, another important muscle homeobox factor, represses differentiation. We now identify Barx2, MyoD, and Pax7 as novel components of the Wnt effector complex, providing a new molecular pathway for regulation of muscle progenitor differentiation. Canonical Wnt signaling induces Barx2 expression in muscle progenitors and perturbation of Barx2 leads to misregulation of Wnt target genes. Barx2 activates two endogenous Wnt target promoters as well as the Wnt reporter gene TOPflash, the latter synergistically with MyoD. Moreover, Barx2 interacts with the core Wnt effectors β-catenin and T cell-factor 4 (TCF4), is recruited to TCF/lymphoid enhancer factor sites, and promotes recruitment of β-catenin. In contrast, Pax7 represses the Wnt reporter gene and antagonizes the activating effect of Barx2. Pax7 also binds β-catenin suggesting that Barx2 and Pax7 may compete for interaction with the core Wnt effector complex. Overall, the data show for the first time that Barx2, Pax7, and MRFs can act as direct transcriptional effectors of Wnt signals in myoblasts and that Barx2 and Wnt signaling participate in a regulatory loop. We propose that antagonism between Barx2 and Pax7 in regulation of Wnt signaling may help mediate the switch from myoblast proliferation to differentiation.
Collapse
Affiliation(s)
- Lizhe Zhuang
- Department of Clinical Pharmacology, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Murphy MM, Keefe AC, Lawson JA, Flygare SD, Yandell M, Kardon G. Transiently active Wnt/β-catenin signaling is not required but must be silenced for stem cell function during muscle regeneration. Stem Cell Reports 2014; 3:475-88. [PMID: 25241745 PMCID: PMC4266007 DOI: 10.1016/j.stemcr.2014.06.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 06/27/2014] [Accepted: 06/30/2014] [Indexed: 12/22/2022] Open
Abstract
Adult muscle’s exceptional capacity for regeneration is mediated by muscle stem cells, termed satellite cells. As with many stem cells, Wnt/β-catenin signaling has been proposed to be critical in satellite cells during regeneration. Using new genetic reagents, we explicitly test in vivo whether Wnt/β-catenin signaling is necessary and sufficient within satellite cells and their derivatives for regeneration. We find that signaling is transiently active in transit-amplifying myoblasts, but is not required for regeneration or satellite cell self-renewal. Instead, downregulation of transiently activated β-catenin is important to limit the regenerative response, as continuous regeneration is deleterious. Wnt/β-catenin activation in adult satellite cells may simply be a vestige of their developmental lineage, in which β-catenin signaling is critical for fetal myogenesis. In the adult, surprisingly, we show that it is not activation but rather silencing of Wnt/β-catenin signaling that is important for muscle regeneration. Wnt/β-catenin signaling is transiently active in myoblasts during muscle regeneration β-catenin is not required in myogenic cells for muscle regeneration β-catenin signaling in myoblasts must be silenced to limit the regenerative response β-catenin requirement and sensitivity differs in fetal and adult muscle stem cells
Collapse
Affiliation(s)
- Malea M Murphy
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra C Keefe
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jennifer A Lawson
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Steven D Flygare
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Mark Yandell
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
42
|
Riganti C, Salaroglio IC, Pinzòn-Daza ML, Caldera V, Campia I, Kopecka J, Mellai M, Annovazzi L, Couraud PO, Bosia A, Ghigo D, Schiffer D. Temozolomide down-regulates P-glycoprotein in human blood-brain barrier cells by disrupting Wnt3 signaling. Cell Mol Life Sci 2014; 71:499-516. [PMID: 23771630 PMCID: PMC11113102 DOI: 10.1007/s00018-013-1397-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 01/08/2023]
Abstract
Low delivery of many anticancer drugs across the blood-brain barrier (BBB) is a limitation to the success of chemotherapy in glioblastoma. This is because of the high levels of ATP-binding cassette transporters like P-glycoprotein (Pgp/ABCB1), which effluxes drugs back to the bloodstream. Temozolomide is one of the few agents able to cross the BBB; its effects on BBB cells permeability and Pgp activity are not known. We found that temozolomide, at therapeutic concentration, increased the transport of Pgp substrates across human brain microvascular endothelial cells and decreased the expression of Pgp. By methylating the promoter of Wnt3 gene, temozolomide lowers the endogenous synthesis of Wnt3 in BBB cells, disrupts the Wnt3/glycogen synthase kinase 3/β-catenin signaling, and reduces the binding of β-catenin on the promoter of mdr1 gene, which encodes for Pgp. In co-culture models of BBB cells and human glioblastoma cells, pre-treatment with temozolomide increases the delivery, cytotoxicity, and antiproliferative effects of doxorubicin, vinblastine, and topotecan, three substrates of Pgp that are usually poorly delivered across BBB. Our work suggests that temozolomide increases the BBB permeability of drugs that are normally effluxed by Pgp back to the bloodstream. These findings may pave the way to new combinatorial chemotherapy schemes in glioblastoma.
Collapse
Affiliation(s)
- Chiara Riganti
- Department of Oncology, University of Turin, Via Santena, 5/bis, 10126, Turin, Italy,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ishikawa M, Iwamoto T, Fukumoto S, Yamada Y. Pannexin 3 inhibits proliferation of osteoprogenitor cells by regulating Wnt and p21 signaling. J Biol Chem 2013; 289:2839-51. [PMID: 24338011 DOI: 10.1074/jbc.m113.523241] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Canonical Wnt signaling and BMP promote the proliferation and differentiation of osteoprogenitors, respectively. However, the regulatory mechanism involved in the transition from proliferation to differentiation is unclear. Here, we show that Panx3 (pannexin 3) plays a key role in this transition by inhibiting the proliferation and promoting the cell cycle exit. Using primary calvarial cells and explants, C3H10T1/2 cells, and C2C12 cells, we found that Panx3 expression inhibited cell growth, whereas the inhibition of endogenous Panx3 expression increased it. We also found that the Panx3 hemichannel inhibited cell growth by promoting β-catenin degradation through GSK3β activation. Additionally, the Panx3 hemichannel inhibited cyclin D1 transcription and Rb phosphorylation through reduced cAMP/PKA/CREB signaling. Furthermore, the Panx3 endoplasmic reticulum Ca(2+) channel induced the transcription and phosphorylation of p21, through the calmodulin/Smad pathway, and resulted in the cell cycle exit. Our results reveal that Panx3 is a new regulator that promotes the switch from proliferation to differentiation of osteoprogenitors via multiple Panx3 signaling pathways.
Collapse
Affiliation(s)
- Masaki Ishikawa
- From the Laboratory of Cell and Developmental Biology, NIDCR, National Institutes of Health, Bethesda, Maryland 20892-4370
| | | | | | | |
Collapse
|
44
|
Interaction of Wnt Signaling with BMP/Smad Signaling during the Transition from Cell Proliferation to Myogenic Differentiation in Mouse Myoblast-Derived Cells. Int J Cell Biol 2013; 2013:616294. [PMID: 23864860 PMCID: PMC3705783 DOI: 10.1155/2013/616294] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 06/03/2013] [Indexed: 11/17/2022] Open
Abstract
Background. Wnt signaling is involved in muscle formation through β-catenin-dependent or -independent pathways, but interactions with other signaling pathways including transforming growth factor β/Smad have not been precisely elucidated. Results. As Wnt4 stimulates myogenic differentiation by antagonizing myostatin (GDF8) activity, we examined the role of Wnt4 signaling during muscle differentiation in the C2C12 myoblast cell line. Among several extrinsic signaling molecules examined in a microarray analysis of C2C12 cells during the transition from cell proliferation to differentiation after mitogen deprivation, bone morphogenetic protein 4 (BMP4) expression was prominently increased. Wnt4 overexpression had similar effects on BMP4 expression. BMP4 was able to inhibit muscle differentiation when added to the culture medium. BMP4 and noggin had no effects on the cellular localization of β-catenin induced by Wnt3a; however, the BMP4-induced phosphorylation of Smad1/5/8 was enhanced by Wnt4, but not by Wnt3a. The BMP antagonist noggin effectively stimulated muscle differentiation through binding to endogenous BMPs, and the effect of noggin was enhanced by the presence of Wnt3a and Wnt4. Conclusion. These results suggest that BMP/Smad pathways are modified through Wnt signaling during the transition from progenitor cell proliferation to myogenic differentiation, although Wnt/β-catenin signaling is not modified with BMP/Smad signaling.
Collapse
|
45
|
Foxc2 induces Wnt4 and Bmp4 expression during muscle regeneration and osteogenesis. Cell Death Differ 2013; 20:1031-42. [PMID: 23645207 DOI: 10.1038/cdd.2013.34] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 03/18/2013] [Accepted: 03/26/2013] [Indexed: 02/06/2023] Open
Abstract
Proliferation and fusion of myoblasts is a well-orchestrated process occurring during muscle development and regeneration. Although myoblasts are known to originate from muscle satellite cells, the molecular mechanisms that coordinate their commitment toward differentiation are poorly understood. Here, we present a novel role for the transcription factor Forkhead box protein C2 (Foxc2) in regulating proliferation and preventing premature differentiation of activated muscle satellite cells. We demonstrate that Foxc2 expression is upregulated early in activated mouse muscle satellite cells and then diminishes during myogenesis. In undifferentiated C2C12 myoblasts, downregulation of endogenous Foxc2 expression leads to a decrease in proliferation, whereas forced expression of FOXC2 sustains proliferation and prevents differentiation into myotubes. We also show that FOXC2 induces Wnt signaling by direct interaction with the Wnt4 (wingless-type MMTV integration site family member-4) promoter region. The resulting elevated expression of bone morphogenetic protein-4 (Bmp4) and RhoA-GTP proteins inhibits the proper myoblast alignment and fusion required for myotube formation. Interestingly, continuous forced expression of FOXC2 alters the commitment of C2C12 myoblasts toward osteogenic differentiation, which is consistent with FOXC2 expression observed in patients with myositis ossificans, an abnormal bone growth within muscle tissue. In summary, our results suggest that (a) Foxc2 regulates the proliferation of multipotent muscle satellite cells; (b) downregulation of Foxc2 is critical for myogenesis to progress; and (c) sustained Foxc2 expression in myoblast cells suppresses myogenesis and alters their lineage commitment toward osteogenesis by inducing the Wnt4 and Bmp4 signaling pathways.
Collapse
|
46
|
M-cadherin-inhibited phosphorylation of ß-catenin augments differentiation of mouse myoblasts. Cell Tissue Res 2012; 351:183-200. [PMID: 23138569 DOI: 10.1007/s00441-012-1515-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 10/05/2012] [Indexed: 10/27/2022]
Abstract
β-Catenin is essential for muscle development because it regulates both cadherin-mediated cell-cell adhesion and canonical Wingless and Int1 (Wnt) signaling. The phosphorylation of β-catenin by glycogen synthase kinase-3β (GSK-3β) at serine31/37/threonine41 regulates its stability and its role in canonical Wnt signaling. In this study, we have investigated whether the N-terminal phosphorylation of β-catenin is regulated by M-cadherin, and whether this regulation mediates the role of M-cadherin in myogenic differentiation. Our data show that the knockdown of M-cadherin expression by RNA interference (RNAi) in C2C12 myoblasts significantly increases the phosphorylation of β-catenin at Ser33/37/Thr41 and decreases the protein abundance of ser37/thr41-unphosphorylated active β-catenin. Furthermore, M-cadherin RNAi promotes TCF/LEF transcription activity but also blunts the initiation of the myogenic progress by Wnt pathway activator lithium chloride or Wnt-3a treatment. Knockdown of β-catenin expression by RNAi decreases myogenic induction in myoblasts. Forced expression of a phosphorylation-resistant β-catenin plasmid (S33Y-β-catenin) fails to enhance myogenic differentiation, but it partially rescues C2C12 cells from M-cadherin RNAi-induced apoptosis. These data show, for the first time, that M-cadherin-mediated signaling attenuates β-catenin phosphorylation at Ser31/37/Thr41 by GSK-3β, and that this regulation has a positive effect on myogenic differentiation induced by canonical Wnt signaling.
Collapse
|
47
|
von Maltzahn J, Chang NC, Bentzinger CF, Rudnicki MA. Wnt signaling in myogenesis. Trends Cell Biol 2012; 22:602-9. [PMID: 22944199 DOI: 10.1016/j.tcb.2012.07.008] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/24/2012] [Accepted: 07/30/2012] [Indexed: 02/05/2023]
Abstract
The formation of skeletal muscle is a tightly regulated process that is critically modulated by Wnt signaling. Myogenesis is dependent on the precise and dynamic integration of multiple Wnt signals allowing self-renewal and progression of muscle precursors in the myogenic lineage. Dysregulation of Wnt signaling can lead to severe developmental defects and perturbation of muscle homeostasis. Recent work has revealed novel roles for the non-canonical planar cell polarity (PCP) and AKT/mTOR pathways in mediating the effects of Wnt on skeletal muscle. In this review, we discuss the role of Wnt signaling in myogenesis and in regulating the homeostasis of adult muscle.
Collapse
Affiliation(s)
- Julia von Maltzahn
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
48
|
Lee JY, Chien IC, Lin WY, Wu SM, Wei BH, Lee YE, Lee HH. Fhl1 as a downstream target of Wnt signaling to promote myogenesis of C2C12 cells. Mol Cell Biochem 2012; 365:251-62. [PMID: 22367176 DOI: 10.1007/s11010-012-1266-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/09/2012] [Indexed: 11/25/2022]
Abstract
Previous studies have shown that Wnt signaling is involved in postnatal mammalian myogenesis; however, the downstream mechanism of Wnt signaling is not fully understood. This study reports that the murine four-and-a-half LIM domain 1 (Fhl1) could be stimulated by β-catenin or LiCl treatment to induce myogenesis. In contrast, knockdown of the Fhl1 gene expression in C2C12 cells led to reduced myotube formation. We also adopted reporter assays to demonstrate that either β-catenin or LiCl significantly activated the Fhl1 promoter, which contains four putative consensus TCF/LEF binding sites. Mutations of two of these sites caused a significant decrease in promoter activity by luciferase reporter assay. Thus, we suggest that Wnt signaling induces muscle cell differentiation, at least partly, through Fhl1 activation.
Collapse
Affiliation(s)
- Jing-Yu Lee
- Department of Bioagricultural Sciences, National Chiayi University, No. 300 Syuefu Rd., Chiayi 60004, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
49
|
Tanaka S, Terada K, Nohno T. Canonical Wnt signaling is involved in switching from cell proliferation to myogenic differentiation of mouse myoblast cells. J Mol Signal 2011; 6:12. [PMID: 21970630 PMCID: PMC3198762 DOI: 10.1186/1750-2187-6-12] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 10/05/2011] [Indexed: 11/29/2022] Open
Abstract
Background Wnt/β-catenin signaling is involved in various aspects of skeletal muscle development and regeneration. In addition, Wnt3a and β-catenin are required for muscle-specific gene transcription in embryonic carcinoma cells and satellite-cell proliferation during adult skeletal muscle regeneration. Downstream targets of canonical Wnt signaling are cyclin D1 and c-myc. However both target genes are suppressed during differentiation of mouse myoblast cells, C2C12. Underlying molecular mechanisms of β-catenin signaling during myogenic differentiation remain unknown. Results Using C2C12 cells, we examined intracellular signaling and gene transcription during myoblast proliferation and differentiation. We confirmed that several Wnt signaling components, including Wnt9a, Sfrp2 and porcupine, were consistently upregulated in differentiating C2C12 cells. Troponin T-positive myotubes were decreased by Wnt3a overexpression, but not Wnt4. TOP/FOP reporter assays revealed that co-expression with Wnt4 reduced Wnt3a-induced luciferase activity, suggesting that Wnt4 signaling counteracted Wnt3a signaling in myoblasts. FH535, a small-molecule inhibitor of β-catenin/Tcf complex formation, reduced basal β-catenin in the cytoplasm and decreased myoblast proliferation. K252a, a protein kinase inhibitor, increased both cytosolic and membrane-bound β-catenin and enhanced myoblast fusion. Treatments with K252a or Wnt4 resulted in increased cytoplasmic vesicles containing phosphorylated β-catenin (Tyr654) during myogenic differentiation. Conclusions These results suggest that various Wnt ligands control subcellular β-catenin localization, which regulate myoblast proliferation and myotube formation. Wnt signaling via β-catenin likely acts as a molecular switch that regulates the transition from cell proliferation to myogenic differentiation.
Collapse
Affiliation(s)
- Shingo Tanaka
- Department of Molecular and Developmental Biology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan.
| | | | | |
Collapse
|