1
|
Chen G, Zou J, He Q, Xia S, Xiao Q, Du R, Zhou S, Zhang C, Wang N, Feng Y. The Role of Non-Coding RNAs in Regulating Cachexia Muscle Atrophy. Cells 2024; 13:1620. [PMID: 39404384 PMCID: PMC11482569 DOI: 10.3390/cells13191620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
Cachexia is a late consequence of various diseases that is characterized by systemic muscle loss, with or without fat loss, leading to significant mortality. Multiple signaling pathways and molecules that increase catabolism, decrease anabolism, and interfere with muscle regeneration are activated. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), play vital roles in cachexia muscle atrophy. This review mainly provides the mechanisms of specific ncRNAs to regulate muscle loss during cachexia and discusses the role of ncRNAs in cachectic biomarkers and novel therapeutic strategies that could offer new insights for clinical practice.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| | - Jiayi Zou
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (Q.H.)
| | - Qianhua He
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (J.Z.); (Q.H.)
| | - Shuyi Xia
- Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
| | - Qili Xiao
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Q.X.); (S.Z.)
| | - Ruoxi Du
- Eighth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China;
| | - Shengmei Zhou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (Q.X.); (S.Z.)
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (G.C.); (C.Z.); (N.W.)
| |
Collapse
|
2
|
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
|
3
|
Tomaz da Silva M, Joshi AS, Castillo MB, Koike TE, Roy A, Gunaratne PH, Kumar A. Fn14 promotes myoblast fusion during regenerative myogenesis. Life Sci Alliance 2023; 6:e202302312. [PMID: 37813488 PMCID: PMC10561765 DOI: 10.26508/lsa.202302312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023] Open
Abstract
Skeletal muscle regeneration involves coordinated activation of an array of signaling pathways. Fibroblast growth factor-inducible 14 (Fn14) is a bona fide receptor for the TWEAK cytokine. Levels of Fn14 are increased in the skeletal muscle of mice after injury. However, the cell-autonomous role of Fn14 in muscle regeneration remains unknown. Here, we demonstrate that global deletion of the Fn14 receptor in mice attenuates muscle regeneration. Conditional ablation of Fn14 in myoblasts but not in differentiated myofibers of mice inhibits skeletal muscle regeneration. Fn14 promotes myoblast fusion without affecting the levels of myogenic regulatory factors in the regenerating muscle. Fn14 deletion in myoblasts hastens initial differentiation but impairs their fusion. The overexpression of Fn14 in myoblasts results in the formation of myotubes having an increased diameter after induction of differentiation. Ablation of Fn14 also reduces the levels of various components of canonical Wnt and calcium signaling both in vitro and in vivo. Forced activation of Wnt signaling rescues fusion defects in Fn14-deficient myoblast cultures. Collectively, our results demonstrate that Fn14-mediated signaling positively regulates myoblast fusion and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Meiricris Tomaz da Silva
- https://ror.org/048sx0r50 Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Aniket S Joshi
- https://ror.org/048sx0r50 Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Micah B Castillo
- https://ror.org/048sx0r50 Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Tatiana E Koike
- https://ror.org/048sx0r50 Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Anirban Roy
- https://ror.org/048sx0r50 Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Preethi H Gunaratne
- https://ror.org/048sx0r50 Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ashok Kumar
- https://ror.org/048sx0r50 Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| |
Collapse
|
4
|
Sharma T, Olea-Flores M, Imbalzano AN. Regulation of the Wnt signaling pathway during myogenesis by the mammalian SWI/SNF ATPase BRG1. Front Cell Dev Biol 2023; 11:1160227. [PMID: 37484913 PMCID: PMC10360407 DOI: 10.3389/fcell.2023.1160227] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Skeletal muscle differentiation is a tightly regulated process, and the importance of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling family for regulation of genes involved in skeletal myogenesis is well-established. Our prior work showed that bromodomains of mSWI/SNF ATPases BRG1 and BRM contribute to myogenesis by facilitating the binding of mSWI/SNF enzymes to regulatory regions of myogenic and other target genes. Here, we report that pathway analyses of differentially expressed genes from that study identified an additional role for mSWI/SNF enzymes via the regulation of the Wnt signaling pathway. The Wnt pathway has been previously shown to be important for skeletal muscle development. To investigate the importance of mSWI/SNF enzymes for the regulation of the Wnt pathway, individual and dual knockdowns were performed for BRG1 and BRM followed by RNA-sequencing. The results show that BRG1, but not BRM, is a regulator of Wnt pathway components and downstream genes. Reactivation of Wnt pathway by stabilization of β-catenin could rescue the defect in myogenic gene expression and differentiation due to BRG1 knockdown or bromodomain inhibition using a specific small molecule inhibitor, PFI-3. These results demonstrate that BRG1 is required upstream of β-catenin function. Chromatin immunoprecipitation of BRG1, BRM and β-catenin at promoters of Wnt pathway component genes showed binding of BRG1 and β-catenin, which provides further mechanistic insight to the transcriptional regulation of these genes.
Collapse
Affiliation(s)
| | | | - Anthony N. Imbalzano
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, United States
| |
Collapse
|
5
|
The Promotion of Migration and Myogenic Differentiation in Skeletal Muscle Cells by Quercetin and Underlying Mechanisms. Nutrients 2022; 14:nu14194106. [PMID: 36235757 PMCID: PMC9572605 DOI: 10.3390/nu14194106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Aging and muscle disorders frequently cause a decrease in myoblast migration and differentiation, leading to losses in skeletal muscle function and regeneration. Several studies have reported that natural flavonoids can stimulate muscle development. Quercetin, one such flavonoid found in many vegetables and fruits, has been used to promote muscle development. In this study, we investigated the effect of quercetin on migration and differentiation, two processes critical to muscle regeneration. We found that quercetin induced the migration and differentiation of mouse C2C12 cells. These results indicated quercetin could induce myogenic differentiation at the early stage through activated p-IGF-1R. The molecular mechanisms of quercetin include the promotion of myogenic differentiation via activated transcription factors STAT3 and the AKT signaling pathway. In addition, we demonstrated that AKT activation is required for quercetin induction of myogenic differentiation to occur. In addition, quercetin was found to promote myoblast migration by regulating the ITGB1 signaling pathway and activating phosphorylation of FAK and paxillin. In conclusion, quercetin can potentially be used to induce migration and differentiation and thus improve muscle regeneration.
Collapse
|
6
|
Posont RJ, Most MS, Cadaret CN, Marks-Nelson ES, Beede KA, Limesand SW, Schmidt TB, Petersen JL, Yates DT. Primary myoblasts from intrauterine growth-restricted fetal sheep exhibit intrinsic dysfunction of proliferation and differentiation that coincides with enrichment of inflammatory cytokine signaling pathways. J Anim Sci 2022; 100:6652330. [PMID: 35908792 PMCID: PMC9339287 DOI: 10.1093/jas/skac145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is linked to lifelong reductions in muscle mass due to intrinsic functional deficits in myoblasts, but the mechanisms underlying these deficits are not known. Our objective was to determine if the deficits were associated with changes in inflammatory and adrenergic regulation of IUGR myoblasts, as was previously observed in IUGR muscle. Primary myoblasts were isolated from IUGR fetal sheep produced by hyperthermia-induced placental insufficiency (PI-IUGR; n = 9) and their controls (n = 9) and from IUGR fetal sheep produced by maternofetal inflammation (MI-IUGR; n = 6) and their controls (n = 7). Proliferation rates were less (P < 0.05) for PI-IUGR myoblasts than their controls and were not affected by incubation with IL-6, TNF-α, norepinephrine, or insulin. IκB kinase inhibition reduced (P < 0.05) proliferation of control myoblasts modestly in basal media but substantially in TNF-α-added media and reduced (P < 0.05) PI-IUGR myoblast proliferation substantially in basal and TNF-α-added media. Proliferation was greater (P < 0.05) for MI-IUGR myoblasts than their controls and was not affected by incubation with TNF-α. Insulin increased (P < 0.05) proliferation in both MI-IUGR and control myoblasts. After 72-h differentiation, fewer (P < 0.05) PI-IUGR myoblasts were myogenin+ than controls in basal and IL-6 added media but not TNF-α-added media. Fewer (P < 0.05) PI-IUGR myoblasts were desmin+ than controls in basal media only. Incubation with norepinephrine did not affect myogenin+ or desmin+ percentages, but insulin increased (P < 0.05) both markers in control and PI-IUGR myoblasts. After 96-h differentiation, fewer (P < 0.05) MI-IUGR myoblasts were myogenin+ and desmin+ than controls regardless of media, although TNF-α reduced (P < 0.05) desmin+ myoblasts for both groups. Differentiated PI-IUGR myoblasts had greater (P < 0.05) TNFR1, ULK2, and TNF-α-stimulated TLR4 gene expression, and PI-IUGR semitendinosus muscle had greater (P < 0.05) TNFR1 and IL6 gene expression, greater (P < 0.05) c-Fos protein, and less (P < 0.05) IκBα protein. Differentiated MI-IUGR myoblasts had greater (P < 0.05) TNFR1 and IL6R gene expression, tended to have greater (P = 0.07) ULK2 gene expression, and had greater (P < 0.05) β-catenin protein and TNF-α-stimulated phosphorylation of NFκB. We conclude that these enriched components of TNF-α/TNFR1/NFκB and other inflammatory pathways in IUGR myoblasts contribute to their dysfunction and help explain impaired muscle growth in the IUGR fetus.
Collapse
Affiliation(s)
- Robert J Posont
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Micah S Most
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Caitlin N Cadaret
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Eileen S Marks-Nelson
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Kristin A Beede
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 65721, USA
| | - Ty B Schmidt
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Jessica L Petersen
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Dustin T Yates
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
7
|
Wang J, Broer T, Chavez T, Zhou CJ, Tran S, Xiang Y, Khodabukus A, Diao Y, Bursac N. Myoblast deactivation within engineered human skeletal muscle creates a transcriptionally heterogeneous population of quiescent satellite-like cells. Biomaterials 2022; 284:121508. [PMID: 35421801 PMCID: PMC9289780 DOI: 10.1016/j.biomaterials.2022.121508] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022]
Abstract
Satellite cells (SCs), the adult Pax7-expressing stem cells of skeletal muscle, are essential for muscle repair. However, in vitro investigations of SC function are challenging due to isolation-induced SC activation, loss of native quiescent state, and differentiation to myoblasts. In the present study, we optimized methods to deactivate in vitro expanded human myoblasts within a 3D culture environment of engineered human skeletal muscle tissues ("myobundles"). Immunostaining and gene expression analyses revealed that a fraction of myoblasts within myobundles adopted a quiescent phenotype (3D-SCs) characterized by increased Pax7 expression, cell cycle exit, and activation of Notch signaling. Similar to native SCs, 3D-SC quiescence is regulated by Notch and Wnt signaling while loss of quiescence and reactivation of 3D-SCs can be induced by growth factors including bFGF. Myobundle injury with a bee toxin, melittin, induces robust myofiber fragmentation, functional decline, and 3D-SC proliferation. By applying single cell RNA-sequencing (scRNA-seq), we discover the existence of two 3D-SC subpopulations (quiescent and activated), identify deactivation-associated gene signature using trajectory inference between 2D myoblasts and 3D-SCs, and characterize the transcriptomic changes within reactivated 3D-SCs in response to melittin-induced injury. These results demonstrate the ability of an in vitro engineered 3D human skeletal muscle environment to support the formation of a quiescent and heterogeneous SC population recapitulating several aspects of the native SC phenotype, and provide a platform for future studies of human muscle regeneration and disease-associated SC dysfunction.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Torie Broer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Ravichandran J, Roust LR, Katsanos CS. Increased Expression of Syncytin-1 in Skeletal Muscle of Humans With Increased Body Mass Index. Front Physiol 2022; 13:858341. [PMID: 35444566 PMCID: PMC9013906 DOI: 10.3389/fphys.2022.858341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/14/2022] [Indexed: 11/23/2022] Open
Abstract
Obesity negatively impacts skeletal muscle protein metabolism, and also impairs skeletal muscle maintenance and regeneration. We analyzed muscle biopsy samples from humans with increased body mass index (BMI) (i.e. > 30 kg/m2) and controls (i.e., BMI < 25 kg/m2) for expression of syncytin-1, a fusogenic protein regulating skeletal muscle regeneration. When compared to controls, humans with increased BMI and concomitant reduction in muscle protein synthesis had higher expression of syncytin-1 in skeletal muscle (p < 0.05). Across human subjects, muscle protein synthesis correlated inversely (r = −0.51; p = 0.03) with syncytin-1 expression in muscle. Using a C2C12 cell line we found that expression of syncytin-A (i.e, corresponding protein in murine tissue) is increased by insulin, and that this response is impaired in the presence of fatty acids, whose metabolism is altered within the metabolic environment induced by increased BMI. In C2C12 cells, the response of the protein 4E-BP1, which signals increase in protein synthesis in muscle, resembled that of syncytin-A. These findings provide novel insights into the expression of syncytin-1 in skeletal muscle of humans with increased BMI, as well as its basic regulation by insulin and fatty acids in muscle. The findings signify the need for further research into the regulation of syncytin-1 in skeletal muscle of humans with increased BMI, as well as its biological implications for altering muscle protein metabolism and regeneration.
Collapse
Affiliation(s)
| | - Lori R. Roust
- College of Medicine, Mayo Clinic in Arizona, Scottsdale, AZ, United States
| | - Christos S. Katsanos
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic in Arizona, Scottsdale, AZ, United States
- *Correspondence: Christos S. Katsanos,
| |
Collapse
|
9
|
Ferrari R, Xie B, Assaf E, Morder K, Scott M, Liao H, Calderon MJ, Ross M, Loughran P, Watkins SC, Pipinos I, Casale G, Tzeng E, McEnaney R, Sachdev U. Inflammatory Caspase Activity Mediates HMGB1 Release and Differentiation in Myoblasts Affected by Peripheral Arterial Disease. Cells 2022; 11:1163. [PMID: 35406727 PMCID: PMC8997414 DOI: 10.3390/cells11071163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/10/2022] Open
Abstract
Introduction: We previously showed that caspase-1 and -11, which are activated by inflammasomes, mediate recovery from muscle ischemia in mice. We hypothesized that similar to murine models, inflammatory caspases modulate myogenicity and inflammation in ischemic muscle disease. Methods: Caspase activity was measured in ischemic and perfused human myoblasts in response to the NLRP3 and AIM2 inflammasome agonists (nigericin and poly(dA:dT), respectively) with and without specific caspase-1 or pan-caspase inhibition. mRNA levels of myogenic markers and caspase-1 were assessed, and protein levels of caspases-1, -4, -5, and -3 were measured by Western blot. Results: When compared to perfused cells, ischemic myoblasts demonstrated attenuated MyoD and myogenin and elevated caspase-1 mRNA. Ischemic myoblasts also had significantly higher enzymatic caspase activity with poly(dA:dT) (p < 0.001), but not nigericin stimulation. Inhibition of caspase activity including caspase-4/-5, but not caspase-1, blocked activation effects of poly(dA:dT). Ischemic myoblasts had elevated cleaved caspase-5. Inhibition of caspase activity deterred differentiation in ischemic but not perfused myoblasts and reduced the release of HMGB1 from both groups. Conclusion: Inflammatory caspases can be activated in ischemic myoblasts by AIM2 and influence ischemic myoblast differentiation and release of pro-angiogenic HMGB1. AIM2 inflammasome involvement suggests a role as a DNA damage sensor, and our data suggest that caspase-5 rather than caspase-1 may mediate the downstream mediator of this pathway.
Collapse
Affiliation(s)
- Ricardo Ferrari
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Bowen Xie
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Edwyn Assaf
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Kristin Morder
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Melanie Scott
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Hong Liao
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| | - Michael J. Calderon
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Mark Ross
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Patricia Loughran
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Simon C. Watkins
- University of Pittsburgh Center for Biologic Imaging, Pittsburgh, PA 15213, USA; (M.J.C.); (M.R.); (P.L.); (S.C.W.)
| | - Iraklis Pipinos
- Department of Surgery, University of Nebraska, Omaha, NE 68198, USA; (I.P.); (G.C.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - George Casale
- Department of Surgery, University of Nebraska, Omaha, NE 68198, USA; (I.P.); (G.C.)
| | - Edith Tzeng
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - Ryan McEnaney
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
- Department of Surgery, Veterans Affairs Hospital, Pittsburgh, PA 15240, USA
| | - Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (R.F.); (B.X.); (E.A.); (K.M.); (M.S.); (H.L.); (E.T.); (R.M.)
| |
Collapse
|
10
|
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
|
11
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
12
|
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
|
13
|
Wang L, Li J, Di LJ. Glycogen synthesis and beyond, a comprehensive review of GSK3 as a key regulator of metabolic pathways and a therapeutic target for treating metabolic diseases. Med Res Rev 2021; 42:946-982. [PMID: 34729791 PMCID: PMC9298385 DOI: 10.1002/med.21867] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/01/2021] [Accepted: 10/24/2021] [Indexed: 12/19/2022]
Abstract
Glycogen synthase kinase‐3 (GSK3) is a highly evolutionarily conserved serine/threonine protein kinase first identified as an enzyme that regulates glycogen synthase (GS) in response to insulin stimulation, which involves GSK3 regulation of glucose metabolism and energy homeostasis. Both isoforms of GSK3, GSK3α, and GSK3β, have been implicated in many biological and pathophysiological processes. The various functions of GSK3 are indicated by its widespread distribution in multiple cell types and tissues. The studies of GSK3 activity using animal models and the observed effects of GSK3‐specific inhibitors provide more insights into the roles of GSK3 in regulating energy metabolism and homeostasis. The cross‐talk between GSK3 and some important energy regulators and sensors and the regulation of GSK3 in mitochondrial activity and component function further highlight the molecular mechanisms in which GSK3 is involved to regulate the metabolic activity, beyond its classical regulatory effect on GS. In this review, we summarize the specific roles of GSK3 in energy metabolism regulation in tissues that are tightly associated with energy metabolism and the functions of GSK3 in the development of metabolic disorders. We also address the impacts of GSK3 on the regulation of mitochondrial function, activity and associated metabolic regulation. The application of GSK3 inhibitors in clinical tests will be highlighted too. Interactions between GSK3 and important energy regulators and GSK3‐mediated responses to different stresses that are related to metabolism are described to provide a brief overview of previously less‐appreciated biological functions of GSK3 in energy metabolism and associated diseases through its regulation of GS and other functions.
Collapse
Affiliation(s)
- Li Wang
- Proteomics, Metabolomics, and Drug Development Core, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Jiajia Li
- Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| | - Li-Jun Di
- Department of Biomedical Sciences, Faculty of Health Sciences, Macau, China.,Cancer Center of the Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, University of Macau, Macau, China.,Ministry of Education, Frontiers Science Center for Precision Oncology, University of Macau, Macau, China
| |
Collapse
|
14
|
Bastin G, Luu L, Batchuluun B, Mighiu A, Beadman S, Zhang H, He C, Al Rijjal D, Wheeler MB, Heximer SP. RGS4-Deficiency Alters Intracellular Calcium and PKA-Mediated Control of Insulin Secretion in Glucose-Stimulated Beta Islets. Biomedicines 2021; 9:biomedicines9081008. [PMID: 34440212 PMCID: PMC8391461 DOI: 10.3390/biomedicines9081008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022] Open
Abstract
A number of diverse G-protein signaling pathways have been shown to regulate insulin secretion from pancreatic β-cells. Accordingly, regulator of G-protein signaling (RGS) proteins have also been implicated in coordinating this process. One such protein, RGS4, is reported to show both positive and negative effects on insulin secretion from β-cells depending on the physiologic context under which it was studied. We here use an RGS4-deficient mouse model to characterize previously unknown G-protein signaling pathways that are regulated by RGS4 during glucose-stimulated insulin secretion from the pancreatic islets. Our data show that loss of RGS4 results in a marked deficiency in glucose-stimulated insulin secretion during both phase I and phase II of insulin release in intact mice and isolated islets. These deficiencies are associated with lower cAMP/PKA activity and a loss of normal calcium surge (phase I) and oscillatory (phase II) kinetics behavior in the RGS4-deficient β-cells, suggesting RGS4 may be important for regulation of both Gαi and Gαq signaling control during glucose-stimulated insulin secretion. Together, these studies add to the known list of G-protein coupled signaling events that are controlled by RGS4 during glucose-stimulated insulin secretion and highlight the importance of maintaining normal levels of RGS4 function in healthy pancreatic tissues.
Collapse
Affiliation(s)
- Guillaume Bastin
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, University of Toronto, Toronto, ON M5G 1M1, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, ON M5S 3H2, Canada
- Correspondence: ; Tel.: +33-658-469-334
| | - Lemieux Luu
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Battsetseg Batchuluun
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Alexandra Mighiu
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Stephanie Beadman
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Hangjung Zhang
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Changhao He
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Dana Al Rijjal
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Michael B. Wheeler
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
| | - Scott P. Heximer
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; (L.L.); (B.B.); (A.M.); (S.B.); (H.Z.); (C.H.); (D.A.R.); (M.B.W.); (S.P.H.)
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, University of Toronto, Toronto, ON M5G 1M1, Canada
- Heart and Stroke/Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
15
|
Key Genes Regulating Skeletal Muscle Development and Growth in Farm Animals. Animals (Basel) 2021; 11:ani11030835. [PMID: 33809500 PMCID: PMC7999090 DOI: 10.3390/ani11030835] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Skeletal muscle mass is an important economic trait, and muscle development and growth is a crucial factor to supply enough meat for human consumption. Thus, understanding (candidate) genes regulating skeletal muscle development is crucial for understanding molecular genetic regulation of muscle growth and can be benefit the meat industry toward the goal of increasing meat yields. During the past years, significant progress has been made for understanding these mechanisms, and thus, we decided to write a comprehensive review covering regulators and (candidate) genes crucial for muscle development and growth in farm animals. Detection of these genes and factors increases our understanding of muscle growth and development and is a great help for breeders to satisfy demands for meat production on a global scale. Abstract Farm-animal species play crucial roles in satisfying demands for meat on a global scale, and they are genetically being developed to enhance the efficiency of meat production. In particular, one of the important breeders’ aims is to increase skeletal muscle growth in farm animals. The enhancement of muscle development and growth is crucial to meet consumers’ demands regarding meat quality. Fetal skeletal muscle development involves myogenesis (with myoblast proliferation, differentiation, and fusion), fibrogenesis, and adipogenesis. Typically, myogenesis is regulated by a convoluted network of intrinsic and extrinsic factors monitored by myogenic regulatory factor genes in two or three phases, as well as genes that code for kinases. Marker-assisted selection relies on candidate genes related positively or negatively to muscle development and can be a strong supplement to classical selection strategies in farm animals. This comprehensive review covers important (candidate) genes that regulate muscle development and growth in farm animals (cattle, sheep, chicken, and pig). The identification of these genes is an important step toward the goal of increasing meat yields and improves meat quality.
Collapse
|
16
|
Grande V, Hathazi D, O'Connor E, Marteau T, Schara-Schmidt U, Hentschel A, Gourdon G, Nikolenko N, Lochmüller H, Roos A. Dysregulation of GSK3β-Target Proteins in Skin Fibroblasts of Myotonic Dystrophy Type 1 (DM1) Patients. J Neuromuscul Dis 2021; 8:603-619. [PMID: 33682722 DOI: 10.3233/jnd-200558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common monogenetic muscular disorder of adulthood. This multisystemic disease is caused by CTG repeat expansion in the 3'-untranslated region of the DM1 protein kinase gene called DMPK. DMPK encodes a myosin kinase expressed in skeletal muscle cells and other cellular populations such as smooth muscle cells, neurons and fibroblasts. The resultant expanded (CUG)n RNA transcripts sequester RNA binding factors leading to ubiquitous and persistent splicing deregulation. The accumulation of mutant CUG repeats is linked to increased activity of glycogen synthase kinase 3 beta (GSK3β), a highly conserved and ubiquitous serine/threonine kinase with functions in pathways regulating inflammation, metabolism, oncogenesis, neurogenesis and myogenesis. As GSK3β-inhibition ameliorates defects in myogenesis, muscle strength and myotonia in a DM1 mouse model, this kinase represents a key player of DM1 pathobiochemistry and constitutes a promising therapeutic target. To better characterise DM1 patients, and monitor treatment responses, we aimed to define a set of robust disease and severity markers linked to GSK3βby unbiased proteomic profiling utilizing fibroblasts derived from DM1 patients with low (80- 150) and high (2600- 3600) CTG-repeats. Apart from GSK3β increase, we identified dysregulation of nine proteins (CAPN1, CTNNB1, CTPS1, DNMT1, HDAC2, HNRNPH3, MAP2K2, NR3C1, VDAC2) modulated by GSK3β. In silico-based expression studies confirmed expression in neuronal and skeletal muscle cells and revealed a relatively elevated abundance in fibroblasts. The potential impact of each marker in the myopathology of DM1 is discussed based on respective function to inform potential uses as severity markers or for monitoring GSK3β inhibitor treatment responses.
Collapse
Affiliation(s)
- Valentina Grande
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany.,Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Emily O'Connor
- Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Theo Marteau
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Ulrike Schara-Schmidt
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Genevieve Gourdon
- Centre de Recherche en Myologie, Association Institut de Myologie, Sorbonne Université, Inserm UMR 974, Paris, France
| | - Nikoletta Nikolenko
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hanns Lochmüller
- Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.,Centro Nacional de AnálisisGenómico, Center for Genomic Regulation (CNAG-CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Andreas Roos
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany.,Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
17
|
Zhang W, Yu L, Han X, Pan J, Deng J, Zhu L, Lu Y, Huang W, Liu S, Li Q, Liu Y. The secretome of human dental pulp stem cells protects myoblasts from hypoxia‑induced injury via the Wnt/β‑catenin pathway. Int J Mol Med 2020; 45:1501-1513. [PMID: 32323739 PMCID: PMC7138287 DOI: 10.3892/ijmm.2020.4525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022] Open
Abstract
Human dental pulp stem cells (hDPSCs) present several advantages, including their ability to be non-invasively harvested without ethical concern. The secretome of hDPSCs can promote the functional recovery of various tissue injuries. However, the protective effects on hypoxia-induced skeletal muscle injury remain to be explored. The present study demonstrated that C2C12 myoblast coculture with hDPSCs attenuated CoCl2-induced hypoxic injury compared with C2C12 alone. The hDPSC secretome increased cell viability and differentiation and decreased G2/M cell cycle arrest under hypoxic conditions. These results were further verified using hDPSC-conditioned medium (hDPSC-CM). The present data revealed that the protective effects of hDPSC-CM depend on the concentration ratio of the CM. In terms of the underlying molecular mechanism, hDPSC-CM activated the Wnt/β-catenin pathway, which increased the protein levels of Wnt1, phosphorylated-glycogen synthase kinase-3β and β-catenin and the mRNA levels of Wnt target genes. By contrast, an inhibitor (XAV939) of Wnt/β-catenin diminished the protective effects of hDPSC-CM. Taken together, the findings of the present study demonstrated that the hDPSC secretome alleviated the hypoxia-induced myoblast injury potentially through regulating the Wnt/β-catenin pathway. These findings may provide new insight into a therapeutic alternative using the hDPSC secretome in skeletal muscle hypoxia-related diseases.
Collapse
Affiliation(s)
- Weihua Zhang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Liming Yu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Xinxin Han
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Jie Pan
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Jiajia Deng
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Luying Zhu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Yun Lu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Wei Huang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Shangfeng Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Qiang Li
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| |
Collapse
|
18
|
Li J, Wang Y, Wang Y, Yan Y, Tong H, Li S. Fibronectin type III domain containing four promotes differentiation of C2C12 through the Wnt/β-catenin signaling pathway. FASEB J 2020; 34:7759-7772. [PMID: 32298013 DOI: 10.1096/fj.201902860rrr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 01/16/2023]
Abstract
Fibronectin type III domain containing 4 (FNDC4) belongs to the fibronectin type III domain containing protein family. FNDC5, which is highly homologous to FNDC4, can promote the differentiation of cardiac cells. We aimed to investigate the role of FNDC4 in the differentiation of C2C12 mouse skeletal muscle cells. Western blotting and immunofluorescence analysis showed that FNDC4 gradually increased with the differentiation of C2C12. Muscle injury repair experiments indicated that FNDC4 may promote the repair of injured muscles. When FNDC4 was either overexpressed or knocked down, the expression of desmin and myogenin myogenic marker molecules followed that of FNDC4, suggesting that FNDC4 can influence the differentiation of C2C12. In addition, immunoprecipitation results showed that FNDC4 can interact with the Wnt/β-catenin signaling pathway receptor low-density lipoprotein receptor-related protein 6 (LRP6), and that β-catenin levels in the nucleus decreased after knocking down FNDC4. Exogenous addition of FNDC4 protein could not restore the blocking of differentiation due to inhibition of both Wnt/β-catenin signal transduction and LRP6 activity via the β-catenin inhibitor XAV-939. Overall, our findings indicate that FDNC4 can influence the differentiation of C2C12 by activating Wnt/β-catenin signal transduction.
Collapse
Affiliation(s)
- Jiwei Li
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yanshuang Wang
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yan Wang
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Yunqin Yan
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Huili Tong
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| | - Shufeng Li
- The Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
19
|
Welc SS, Wehling-Henricks M, Kuro-o M, Thomas KA, Tidball JG. Modulation of Klotho expression in injured muscle perturbs Wnt signalling and influences the rate of muscle growth. Exp Physiol 2020; 105:132-147. [PMID: 31724771 PMCID: PMC6938556 DOI: 10.1113/ep088142] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does modulating the expression of Klotho affect myogenesis following acute injury of healthy, non-senescent muscle? What is the main finding and its importance? Klotho can accelerate muscle growth following acute injury of healthy, adult mice, which supports the possibility that increased delivery of Klotho could have therapeutic value for improving repair of damaged muscle. ABSTRACT Skeletal muscle injuries activate a complex programme of myogenesis that can restore normal muscle structure. We tested whether modulating the expression of klotho influenced the response of mouse muscles to acute injury. Our findings show that klotho expression in muscle declines at 3 days post-injury. That reduction in klotho expression coincided with elevated expression of targets of Wnt signalling (Ccnd1; Myc) and increased MyoD+ muscle cell numbers, reflecting the onset of myogenic cell differentiation. klotho expression subsequently increased at 7 days post-injury with elevated expression occurring primarily in inflammatory lesions, which was accompanied by reduced expression of Wnt target genes (Ccnd1: 91%; Myc: 96%). Introduction of a klotho transgene maintained high levels of klotho expression over the course of muscle repair and attenuated the increases in Ccnd1 and Myc expression that occurred at 3 days post-injury. Correspondingly, transgene expression reduced Wnt signalling in Pax7+ cells, reflected by reductions in Pax7+ cells expressing active β-catenin, and reduced the numbers of MyoD+ cells at 3 days post-injury. At 21 days post-injury, muscles in klotho transgenic mice showed increased Pax7+ and decreased myogenin+ cell densities and large increases in myofibre size. Likewise, treating myogenic cells in vitro with Klotho reduced Myod expression but did not affect Pax7 expression. Muscle inflammation was only slightly modulated by increased klotho expression, initially reducing the expression of M2-biased macrophage markers Cd163 and Cd206 at 3 days post-injury and later increasing the expression of pan-macrophage marker F480 and Cd68 at 21 days post-injury. Collectively, our study shows that Klotho modulates myogenesis and that increased expression accelerates muscle growth after injury.
Collapse
Affiliation(s)
- Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Indiana University School of Medicine, 635 Barnhill Drive, MS-332, Indianapolis, IN 46202
| | | | - Makoto Kuro-o
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Kyle A. Thomas
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - James G. Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA
| |
Collapse
|
20
|
Davuluri G, Giusto M, Chandel R, Welch N, Alsabbagh K, Kant S, Kumar A, Kim A, Gangadhariah M, Ghosh PK, Tran U, Krajcik DM, Vasu K, DiDonato AJ, DiDonato JA, Willard B, Monga SP, Wang Y, Fox PL, Stark GR, Wessely O, Esser KA, Dasarathy S. Impaired Ribosomal Biogenesis by Noncanonical Degradation of β-Catenin during Hyperammonemia. Mol Cell Biol 2019; 39:e00451-18. [PMID: 31138664 PMCID: PMC6664607 DOI: 10.1128/mcb.00451-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/19/2018] [Accepted: 05/18/2019] [Indexed: 12/21/2022] Open
Abstract
Increased ribosomal biogenesis occurs during tissue hypertrophy, but whether ribosomal biogenesis is impaired during atrophy is not known. We show that hyperammonemia, which occurs in diverse chronic disorders, impairs protein synthesis as a result of decreased ribosomal content and translational capacity. Transcriptome analyses, real-time PCR, and immunoblotting showed consistent reductions in the expression of the large and small ribosomal protein subunits (RPL and RPS, respectively) in hyperammonemic murine skeletal myotubes, HEK cells, and skeletal muscle from hyperammonemic rats and human cirrhotics. Decreased ribosomal content was accompanied by decreased expression of cMYC, a positive regulator of ribosomal biogenesis, as well as reduced expression and activity of β-catenin, a transcriptional activator of cMYC. However, unlike the canonical regulation of β-catenin via glycogen synthase kinase 3β (GSK3β)-dependent degradation, GSK3β expression and phosphorylation were unaltered during hyperammonemia, and depletion of GSK3β did not prevent ammonia-induced degradation of β-catenin. Overexpression of GSK3β-resistant variants, genetic depletion of IκB kinase β (IKKβ) (activated during hyperammonemia), protein interactions, and in vitro kinase assays showed that IKKβ phosphorylated β-catenin directly. Overexpressing β-catenin restored hyperammonemia-induced perturbations in signaling responses that regulate ribosomal biogenesis. Our data show that decreased protein synthesis during hyperammonemia is mediated via a novel GSK3β-independent, IKKβ-dependent impairment of the β-catenin-cMYC axis.
Collapse
Affiliation(s)
- Gangarao Davuluri
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Michela Giusto
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rajeev Chandel
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicole Welch
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Khaled Alsabbagh
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sashi Kant
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Avinash Kumar
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Adam Kim
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Prabar K Ghosh
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Uyen Tran
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Daniel M Krajcik
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kommireddy Vasu
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Anthony J DiDonato
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Joseph A DiDonato
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Belinda Willard
- Proteomics Research Core Services, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yuxin Wang
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paul L Fox
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - George R Stark
- Department of Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Oliver Wessely
- Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, Institute of Myology, University of Florida, Gainesville, Florida, USA
| | - Srinivasan Dasarathy
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
21
|
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
|
22
|
Saleh A, Subramaniam G, Raychaudhuri S, Dhawan J. Cytoplasmic sequestration of the RhoA effector mDiaphanous1 by Prohibitin2 promotes muscle differentiation. Sci Rep 2019; 9:8302. [PMID: 31165762 PMCID: PMC6549159 DOI: 10.1038/s41598-019-44749-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscle differentiation is controlled by adhesion and growth factor-dependent signalling through common effectors that regulate muscle-specific transcriptional programs. Here we report that mDiaphanous1, an effector of adhesion-dependent RhoA-signalling, negatively regulates myogenesis at the level of Myogenin expression. In myotubes, over-expression of mDia1ΔN3, a RhoA-independent mutant, suppresses Myogenin promoter activity and expression. We investigated mDia1-interacting proteins that may counteract mDia1 to permit Myogenin expression and timely differentiation. Using yeast two-hybrid and mass-spectrometric analysis, we report that mDia1 has a stage-specific interactome, including Prohibitin2, MyoD, Akt2, and β-Catenin, along with a number of proteosomal and mitochondrial components. Of these interacting partners, Prohibitin2 colocalises with mDia1 in cytoplasmic punctae in myotubes. We mapped the interacting domains of mDia1 and Phb2, and used interacting (mDia1ΔN3/Phb2 FL or mDia1ΔN3/Phb2-Carboxy) and non-interacting pairs (mDia1H + P/Phb2 FL or mDia1ΔN3/Phb2-Amino) to dissect the functional consequences of this partnership on Myogenin promoter activity. Co-expression of full-length as well as mDia1-interacting domains of Prohibitin2 reverse the anti-myogenic effects of mDia1ΔN3, while non-interacting regions do not. Our results suggest that Prohibitin2 sequesters mDia1, dampens its anti-myogenic activity and fine-tunes RhoA-mDia1 signalling to promote differentiation. Overall, we report that mDia1 is multi-functional signalling effector whose anti-myogenic activity is modulated by a differentiation-dependent interactome. The data have been deposited to the ProteomeXchange with identifier PXD012257.
Collapse
Affiliation(s)
- Amena Saleh
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gunasekaran Subramaniam
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Swasti Raychaudhuri
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Science & Regenerative Medicine, Bangalore, Karnataka, 560065, India.
- Council of Scientific & Industrial Research -Centre for Cellular & Molecular Biology, Hyderabad, Telangana, 500007, India.
| |
Collapse
|
23
|
Raslan AA, Yoon JK. R-spondins: Multi-mode WNT signaling regulators in adult stem cells. Int J Biochem Cell Biol 2019; 106:26-34. [DOI: 10.1016/j.biocel.2018.11.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/04/2018] [Accepted: 11/09/2018] [Indexed: 01/08/2023]
|
24
|
Redshaw Z, Loughna PT. Adipogenic Differentiation of Muscle Derived Cells is Repressed by Inhibition of GSK-3 Activity. Front Vet Sci 2018; 5:110. [PMID: 29946551 PMCID: PMC6005818 DOI: 10.3389/fvets.2018.00110] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/04/2018] [Indexed: 12/25/2022] Open
Abstract
Intramuscular fat is important in large animal livestock species in regard to meat quality and in humans is of clinical significance in particular in relation to insulin resistance. The canonical Wnt signalling pathway has been implicated at a whole body level in regulating relative levels of adiposity versus lean body mass. Previously we have shown that pig muscle cells can undergo adipogenic differentiation to a degree that is dependent upon the specific muscle source. In this work we examine the role of the canonical Wnt pathway which acts through inactivation of glycogen synthase kinase-3 (GSK-3) in the regulation of adipogenic differentiation in muscle cells derived from the pig semimembranosus muscle. The application of lithium chloride to muscle derived cells significantly increased the phosphorylation of GSK-3β and thus inhibited its activity thus mimicking Wnt signaling. This was associated with a significant decrease in the expression of the adipogenic transcription factor PPARγ and an almost complete inhibition of adipogenesis in the cells. The data also suggest that GSK-3α plays, at most, a small role in this process. Studies in vivo have suggested that the Wnt pathway is a major regulator of whole body adiposity. In this study we have shown that the ability of cells derived from porcine skeletal muscle to differentiate along an adipogenic lineage, in vitro, is severely impaired by mimicking the action of this pathway. This was done by inactivation of GSK-3β by the use of Lithium Chloride.
Collapse
Affiliation(s)
- Zoe Redshaw
- School of Veterinary Medicine and Science, The University of Nottingham, Loughborough, United Kingdom.,Faculty of Health and Life Sciences, De Montfort University, Leicester, United Kingdom
| | - Paul Thomas Loughna
- School of Veterinary Medicine and Science, The University of Nottingham, Loughborough, United Kingdom
| |
Collapse
|
25
|
FoxO1: a novel insight into its molecular mechanisms in the regulation of skeletal muscle differentiation and fiber type specification. Oncotarget 2018; 8:10662-10674. [PMID: 27793012 PMCID: PMC5354690 DOI: 10.18632/oncotarget.12891] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/19/2016] [Indexed: 02/03/2023] Open
Abstract
FoxO1, a member of the forkhead transcription factor forkhead box protein O (FoxO) family, is predominantly expressed in most muscle types. FoxO1 is a key regulator of muscle growth, metabolism, cell proliferation and differentiation. In the past two decades, many researches have indicated that FoxO1 is a negative regulator of skeletal muscle differentiation while contrasting opinions consider that FoxO1 is crucial for myoblast fusion. FoxO1 is expressed much higher in fast twitch fiber enriched muscles than in slow muscles and is also closely related to muscle fiber type specification. In this review, we summarize the molecular mechanisms of FoxO1 in the regulation of skeletal muscle differentiation and fiber type specification.
Collapse
|
26
|
Kneppers A, Verdijk L, de Theije C, Corten M, Gielen E, van Loon L, Schols A, Langen R. A novel in vitro model for the assessment of postnatal myonuclear accretion. Skelet Muscle 2018; 8:4. [PMID: 29444710 PMCID: PMC5813369 DOI: 10.1186/s13395-018-0151-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 01/26/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Due to the post-mitotic nature of myonuclei, postnatal myogenesis is essential for skeletal muscle growth, repair, and regeneration. This process is facilitated by satellite cells through proliferation, differentiation, and subsequent fusion with a pre-existing muscle fiber (i.e., myonuclear accretion). Current knowledge of myogenesis is primarily based on the in vitro formation of syncytia from myoblasts, which represents aspects of developmental myogenesis, but may incompletely portray postnatal myogenesis. Therefore, we aimed to develop an in vitro model that better reflects postnatal myogenesis, to study the cell intrinsic and extrinsic processes and signaling involved in the regulation of postnatal myogenesis. METHODS Proliferating C2C12 myoblasts were trypsinized and co-cultured for 3 days with 5 days differentiated C2C12 myotubes. Postnatal myonuclear accretion was visually assessed by live cell time-lapse imaging and cell tracing by cell labeling with Vybrant® DiD and DiO. Furthermore, a Cre/LoxP-based cell system was developed to semi-quantitatively assess in vitro postnatal myonuclear accretion by the conditional expression of luciferase upon myoblast-myotube fusion. Luciferase activity was assessed luminometrically and corrected for total protein content. RESULTS Live cell time-lapse imaging, staining-based cell tracing, and recombination-dependent luciferase activity, showed the occurrence of postnatal myonuclear accretion in vitro. Treatment of co-cultures with the myogenic factor IGF-I (p < 0.001) and the cytokines IL-13 (p < 0.05) and IL-4 (p < 0.001) increased postnatal myonuclear accretion, while the myogenic inhibitors cytochalasin D (p < 0.001), myostatin (p < 0.05), and TNFα (p < 0.001) decreased postnatal myonuclear accretion. Furthermore, postnatal myonuclear accretion was increased upon recovery from electrical pulse stimulation-induced fiber damage (p < 0.001) and LY29004-induced atrophy (p < 0.001). Moreover, cell type-specific siRNA-mediated knockdown of myomaker in myoblasts (p < 0.001), but not in myotubes, decreased postnatal myonuclear accretion. CONCLUSIONS We developed a physiologically relevant, sensitive, high-throughput cell system for semi-quantitative assessment of in vitro postnatal myonuclear accretion, which can be used to mimic physiological myogenesis triggers, and can distinguish the cell type-specific roles of signals and responses in the regulation of postnatal myogenesis. As such, this method is suitable for both basal and translational research on the regulation of postnatal myogenesis, and will improve our understanding of muscle pathologies that result from impaired satellite cell number or function.
Collapse
Affiliation(s)
- Anita Kneppers
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Lex Verdijk
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Chiel de Theije
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Mark Corten
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ellis Gielen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Luc van Loon
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Annemie Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ramon Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
27
|
Park KS, Mitra A, Rahat B, Kim K, Pfeifer K. Loss of imprinting mutations define both distinct and overlapping roles for misexpression of IGF2 and of H19 lncRNA. Nucleic Acids Res 2018; 45:12766-12779. [PMID: 29244185 PMCID: PMC5727439 DOI: 10.1093/nar/gkx896] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/26/2017] [Indexed: 12/26/2022] Open
Abstract
Imprinted genes occur in discrete clusters that are coordinately regulated by shared DNA elements called Imprinting Control Regions. H19 and Igf2 are linked imprinted genes that play critical roles in development. Loss of imprinting (LOI) at the IGF2/H19 locus on the maternal chromosome is associated with the developmental disorder Beckwith Wiedemann Syndrome (BWS) and with several cancers. Here we use comprehensive genetic and genomic analyses to follow muscle development in a mouse model of BWS to dissect the separate and shared roles for misexpression of Igf2 and H19 in the disease phenotype. We show that LOI results in defects in muscle differentiation and hypertrophy and identify primary downstream targets: Igf2 overexpression results in over-activation of MAPK signaling while loss of H19 lncRNA prevents normal down regulation of p53 activity and therefore results in reduced AKT/mTOR signaling. Moreover, we demonstrate instances where H19 and Igf2 misexpression work separately, cooperatively, and antagonistically to establish the developmental phenotype. This study thus identifies new biochemical roles for the H19 lncRNA and underscores that LOI phenotypes are multigenic so that complex interactions will contribute to disease outcomes.
Collapse
Affiliation(s)
- Ki-Sun Park
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| | - Apratim Mitra
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| | - Beenish Rahat
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| | - Keekwang Kim
- Department of Biochemistry, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Karl Pfeifer
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
28
|
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]
|
29
|
Hindi SM, Shin J, Gallot YS, Straughn AR, Simionescu-Bankston A, Hindi L, Xiong G, Friedland RP, Kumar A. MyD88 promotes myoblast fusion in a cell-autonomous manner. Nat Commun 2017; 8:1624. [PMID: 29158520 PMCID: PMC5696367 DOI: 10.1038/s41467-017-01866-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 10/20/2017] [Indexed: 12/27/2022] Open
Abstract
Myoblast fusion is an indispensable step for skeletal muscle development, postnatal growth, and regeneration. Myeloid differentiation primary response gene 88 (MyD88) is an adaptor protein that mediates Toll-like receptors and interleukin-1 receptor signaling. Here we report a cell-autonomous role of MyD88 in the regulation of myoblast fusion. MyD88 protein levels are increased during in vitro myogenesis and in conditions that promote skeletal muscle growth in vivo. Deletion of MyD88 impairs fusion of myoblasts without affecting their survival, proliferation, or differentiation. MyD88 regulates non-canonical NF-κB and canonical Wnt signaling during myogenesis and promotes skeletal muscle growth and overload-induced myofiber hypertrophy in mice. Ablation of MyD88 reduces myofiber size during muscle regeneration, whereas its overexpression promotes fusion of exogenous myoblasts to injured myofibers. Our study shows that MyD88 modulates myoblast fusion and suggests that augmenting its levels may be a therapeutic approach to improve skeletal muscle formation in degenerative muscle disorders.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Jonghyun Shin
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Yann S Gallot
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Alex R Straughn
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Adriana Simionescu-Bankston
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Lubna Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Guangyan Xiong
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Robert P Friedland
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
30
|
Agley CC, Lewis FC, Jaka O, Lazarus NR, Velloso C, Francis-West P, Ellison-Hughes GM, Harridge SDR. Active GSK3β and an intact β-catenin TCF complex are essential for the differentiation of human myogenic progenitor cells. Sci Rep 2017; 7:13189. [PMID: 29030569 PMCID: PMC5640663 DOI: 10.1038/s41598-017-10731-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/14/2017] [Indexed: 01/14/2023] Open
Abstract
Wnt-β-catenin signalling is essential for skeletal muscle myogenesis during development, but its role in adult human skeletal muscle remains unknown. Here we have used human primary CD56Pos satellite cell-derived myogenic progenitors obtained from healthy individuals to study the role of Wnt-β-catenin signalling in myogenic differentiation. We show that dephosphorylated β-catenin (active-β-catenin), the central effector of the canonical Wnt cascade, is strongly upregulated at the onset of differentiation and undergoes nuclear translocation as differentiation progresses. To establish the role of Wnt signalling in regulating the differentiation process we manipulated key nodes of this pathway through a series of β-catenin gain-of-function (GSK3 inhibition and β-catenin overexpression) or loss-of-function experiments (dominant negative TCF4). Our data showed that manipulation of these critical pathway components led to varying degrees of disruption to the normal differentiation phenotype indicating the importance of Wnt signalling in regulating this process. We reveal an independent necessity for active-β-catenin in the fusion and differentiation of human myogenic progenitors and that dominant negative inhibition of TCF4 prevents differentiation completely. Together these data add new mechanistic insights into both Wnt signalling and adult human myogenic progenitor differentiation.
Collapse
Affiliation(s)
- C C Agley
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK. .,Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - F C Lewis
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| | - O Jaka
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - N R Lazarus
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - C Velloso
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK
| | - P Francis-West
- Department of Craniofacial development and stem cell biology, King's College London, London, UK
| | - G M Ellison-Hughes
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| | - S D R Harridge
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, UK.,Stem Cell Institute, King's College London, London, UK
| |
Collapse
|
31
|
Effects of genetic variants of the bovine WNT8A gene on nine important growth traits in beef cattle. J Genet 2017; 96:535-544. [PMID: 28947701 DOI: 10.1007/s12041-017-0804-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
WNT-β-catenin-TCF pathway is involved in carcinogenesis and foetal development. As a member of the WNT gene family, Wnt8A encodes secreted signalling proteins and responds to many biological processes.However, similar research on the effects of genetic variations of Wnt8A gene on growth traits is lacking. Therefore, in this study, polymorphisms of Wnt8A were detected in 396 animals from Chinese Qinchuan cattle using DNA pool sequencing and PCR-RFLP methods. Four novel single-nucleotide polymorphisms (SNPs) of Wnt8A gene were identified, including three mutations in introns (g.T-445C, g.G244C and g.G910A) and one in exon (g.T4922C). Additionally, we examined the associations of four SNPs with growth traits. The results revealed that SNP2 (g.G244C) was significantly associated with shoulder height, hip height, body length, hip width, and body weight (P < 0.05). SNP3 (g.G910A) also displayed notable effects on hip width (P < 0.05). Meanwhile, the haplotype combination CC-GC-GA-CC was strongly associated with heavier, taller and longer animals (P < 0.05). These results show that the Wnt8A gene may be a potential candidate gene, and the SNPs could be used as molecular markers in early marker-assisted selection in beef cattle breeding programmes.
Collapse
|
32
|
Preclinical testing of the glycogen synthase kinase-3β inhibitor tideglusib for rhabdomyosarcoma. Oncotarget 2017; 8:62976-62983. [PMID: 28968964 PMCID: PMC5609896 DOI: 10.18632/oncotarget.18520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 06/01/2017] [Indexed: 12/21/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common childhood soft tissue sarcoma. RMS often arise from myogenic precursors and displays a poorly differentiated skeletal muscle phenotype most closely resembling regenerating muscle. GSK3β is a ubiquitously expressed serine-threonine kinase capable of repressing the terminal myogenic differentiation program in cardiac and skeletal muscle. Recent unbiased chemical screening efforts have prioritized GSK3β inhibitors as inducers of myodifferentiation in RMS, suggesting efficacy as single agents in suppressing growth and promoting self-renewal in zebrafish transgenic embryonal RMS (eRMS) models in vivo. In this study, we tested the irreversible GSK3β-inhibitor, tideglusib for in vivo efficacy in patient-derived xenograft models of both alveolar rhabdomyosarcoma (aRMS) and eRMS. Tideglusib had effective on-target pharmacodynamic efficacy, but as a single agent had no effect on tumor progression or myodifferentiation. These results suggest that as monotherapy, GSK3β inhibitors may not be a viable treatment for aRMS or eRMS.
Collapse
|
33
|
Genovese NJ, Domeier TL, Telugu BPVL, Roberts RM. Enhanced Development of Skeletal Myotubes from Porcine Induced Pluripotent Stem Cells. Sci Rep 2017; 7:41833. [PMID: 28165492 PMCID: PMC5292944 DOI: 10.1038/srep41833] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/30/2016] [Indexed: 02/07/2023] Open
Abstract
The pig is recognized as a valuable model in biomedical research in addition to its agricultural importance. Here we describe a means for generating skeletal muscle efficiently from porcine induced pluripotent stem cells (piPSC) in vitro thereby providing a versatile platform for applications ranging from regenerative biology to the ex vivo cultivation of meat. The GSK3B inhibitor, CHIR99021 was employed to suppress apoptosis, elicit WNT signaling events and drive naïve-type piPSC along the mesoderm lineage, and, in combination with the DNA methylation inhibitor 5-aza-cytidine, to activate an early skeletal muscle transcription program. Terminal differentiation was then induced by activation of an ectopically expressed MYOD1. Myotubes, characterized by myofibril development and both spontaneous and stimuli-elicited excitation-contraction coupling cycles appeared within 11 days. Efficient lineage-specific differentiation was confirmed by uniform NCAM1 and myosin heavy chain expression. These results provide an approach for generating skeletal muscle that is potentially applicable to other pluripotent cell lines and to generating other forms of muscle.
Collapse
Affiliation(s)
- Nicholas J Genovese
- C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 6521, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65211, USA
| | - Bhanu Prakash V L Telugu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA.,Animal Bioscience and Biotechnology Laboratory, USDA ARS, Beltsville, MD 20705, USA
| | - R Michael Roberts
- C.S. Bond Life Sciences Center, University of Missouri, Columbia, MO 6521, USA
| |
Collapse
|
34
|
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
|
35
|
Abraham ST. A role for the Wnt3a/β-catenin signaling pathway in the myogenic program of C2C12 cells. In Vitro Cell Dev Biol Anim 2016; 52:935-941. [DOI: 10.1007/s11626-016-0058-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/09/2016] [Indexed: 11/27/2022]
|
36
|
|
37
|
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
|
38
|
BAMBI Promotes C2C12 Myogenic Differentiation by Enhancing Wnt/β-Catenin Signaling. Int J Mol Sci 2015; 16:17734-45. [PMID: 26247931 PMCID: PMC4581218 DOI: 10.3390/ijms160817734] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 07/07/2015] [Accepted: 07/30/2015] [Indexed: 12/03/2022] Open
Abstract
Bone morphogenic protein and activin membrane-bound inhibitor (BAMBI) is regarded as an essential regulator of cell proliferation and differentiation that represses transforming growth factor-β and enhances Wnt/β-catenin signaling in various cell types. However, its role in skeletal muscle remains largely unknown. In the current study, we found that the expression level of BAMBI peaked in the early differentiation phase of the C2C12 rodent myoblast cell line. Knockdown of BAMBI via siRNA inhibited C2C12 differentiation, indicated by repressed MyoD, MyoG, and MyHC expression as well as reductions in the differentiation and fusion indices. BAMBI knockdown reduced the activity of Wnt/β-catenin signaling, as characterized by the decreased nuclear translocation of β-catenin and the lowered transcription of Axin2, which is a well-documented target gene of the Wnt/β-catenin signaling pathway. Furthermore, treatment with LiCl, an activator of Wnt/β-catenin signaling, rescued the reduction in C2C12 differentiation caused by BAMBI siRNA. Taken together, our data suggest that BAMBI is required for normal C2C12 differentiation, and that its role in myogenesis is mediated by the Wnt/β-catenin pathway.
Collapse
|
39
|
Li Q, Li Y, Gu B, Fang L, Zhou P, Bao S, Huang L, Dai X. Akt Phosphorylates Wnt Coactivator and Chromatin Effector Pygo2 at Serine 48 to Antagonize Its Ubiquitin/Proteasome-mediated Degradation. J Biol Chem 2015; 290:21553-67. [PMID: 26170450 DOI: 10.1074/jbc.m115.639419] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Indexed: 01/16/2023] Open
Abstract
Pygopus 2 (Pygo2/PYGO2) is an evolutionarily conserved coactivator and chromatin effector in the Wnt/β-catenin signaling pathway that regulates cell growth and differentiation in various normal and malignant tissues. Although PYGO2 is highly overexpressed in a number of human cancers, the molecular mechanism underlying its deregulation is largely unknown. Here we report that Pygo2 protein is degraded through the ubiquitin/proteasome pathway and is posttranslationally stabilized through phosphorylation by activated phosphatidylinositol 3-kinase/Akt signaling. Specifically, Pygo2 is stabilized upon inhibition of the proteasome, and its intracellular level is regulated by Cullin 4 (Cul4) and DNA damage-binding protein 1 (DDB1), components of the Cul4-DDB1 E3 ubiquitin ligase complex. Furthermore, Pygo2 is phosphorylated at multiple residues, and Akt-mediated phosphorylation at serine 48 leads to its decreased ubiquitylation and increased stability. Finally, we provide evidence that Akt and its upstream growth factors act in parallel with Wnt to stabilize Pygo2. Taken together, our findings highlight chromatin regulator Pygo2 as a common node downstream of oncogenic Wnt and Akt signaling pathways and underscore posttranslational modification, particularly phosphorylation and ubiquitylation, as a significant mode of regulation of Pygo2 protein expression.
Collapse
Affiliation(s)
- Qiuling Li
- From the Department of Biological Chemistry, the State Key Laboratory of Molecular and Developmental Biology, Center for Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China, and
| | - Yuewei Li
- the Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10065
| | - Bingnan Gu
- From the Department of Biological Chemistry
| | - Lei Fang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697
| | - Pengbo Zhou
- the Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10065
| | - Shilai Bao
- the State Key Laboratory of Molecular and Developmental Biology, Center for Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China, and
| | - Lan Huang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697,
| | - Xing Dai
- From the Department of Biological Chemistry,
| |
Collapse
|
40
|
Insulin Effects on Survival of Human Multiple Myeloma Cells. Bull Exp Biol Med 2015; 159:262-5. [PMID: 26087753 DOI: 10.1007/s10517-015-2937-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Indexed: 10/23/2022]
Abstract
Insulin effects of human multiple myeloma cell survival were studied on RPMI1640, RPMI8226, and IM9 lines differing by differentiation degree. The effects of exogenous insulin on tumor cell growth and survival varied. Insulin alone did not improve the viability of myeloma cells, while in combination with serum growth factors increased it. The IM9 cells with immunophenotype (CD(138+), CD(38-), CD(45+), CD(56-), CD(19+)) exhibited the highest sensitivity to serum growth factors, while RPMI1640 and RPMI8226 cells with (CD(138+), CD(38+), CD(45-), CD(56±), CD(19-)) immunophenotype were less sensitive. Studies of gene expression showed a significantly lower level of IRA mRNA expression in IM9 vs. RPMI1640 and RPMI8226 cells.
Collapse
|
41
|
Jones AE, Price FD, Le Grand F, Soleimani VD, Dick SA, Megeney LA, Rudnicki MA. Wnt/β-catenin controls follistatin signalling to regulate satellite cell myogenic potential. Skelet Muscle 2015; 5:14. [PMID: 25949788 PMCID: PMC4421991 DOI: 10.1186/s13395-015-0038-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/08/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Adult skeletal muscle regeneration is a highly orchestrated process involving the activation and proliferation of satellite cells, an adult skeletal muscle stem cell. Activated satellite cells generate a transient amplifying progenitor pool of myoblasts that commit to differentiation and fuse into multinucleated myotubes. During regeneration, canonical Wnt signalling is activated and has been implicated in regulating myogenic lineage progression and terminal differentiation. METHODS Here, we have undertaken a gene expression analysis of committed satellite cell-derived myoblasts to examine their ability to respond to canonical Wnt/β-catenin signalling. RESULTS We found that activation of canonical Wnt signalling induces follistatin expression in myoblasts and promotes myoblast fusion in a follistatin-dependent manner. In growth conditions, canonical Wnt/β-catenin signalling prime myoblasts for myogenic differentiation by stimulating myogenin and follistatin expression. We further found that myogenin binds elements in the follistatin promoter and thus acts downstream of myogenin during differentiation. Finally, ectopic activation of canonical Wnt signalling in vivo promoted premature differentiation during muscle regeneration following acute injury. CONCLUSIONS Together, these data reveal a novel mechanism by which myogenin mediates the canonical Wnt/β-catenin-dependent activation of follistatin and induction of the myogenic differentiation process.
Collapse
Affiliation(s)
- Andrew E Jones
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| | - Feodor D Price
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| | - Fabien Le Grand
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), 24 Rue du Fg St Jacques, Paris, France
| | - Vahab D Soleimani
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada
| | - Sarah A Dick
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| | - Lynn A Megeney
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| | - Michael A Rudnicki
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6 Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| |
Collapse
|
42
|
Figeac N, Zammit PS. Coordinated action of Axin1 and Axin2 suppresses β-catenin to regulate muscle stem cell function. Cell Signal 2015; 27:1652-65. [PMID: 25866367 DOI: 10.1016/j.cellsig.2015.03.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/23/2015] [Indexed: 01/01/2023]
Abstract
The resident stem cells of skeletal muscle are satellite cells, which are regulated by both canonical and non-canonical Wnt pathways. Canonical Wnt signalling promotes differentiation, and is controlled at many levels, including via Axin1 and Axin2-mediated β-catenin degradation. Axin1 and Axin2 are thought equivalent suppressors of canonical Wnt signalling, although Axin2 is also a Wnt target gene. We show that Axin1 expression was higher in proliferating satellite cells, while Axin2 was up-regulated during differentiation. siRNA-mediated Axin1 knockdown changed cell morphology, suppressed proliferation and promoted myogenic differentiation. Simultaneous knockdown of both Axin1 and β-catenin rescued proliferation and partially, premature differentiation. Surprisingly, retroviral-mediated overexpression of Axin2 was unable to compensate for knockdown of Axin1 in satellite cells, indicating that Axin1 and Axin2 are not fully redundant. Isolated satellite cells from Axin2-null mice also had no major phenotype. However, siRNA-mediated knockdown of Axin1 in Axin2-null cells strongly inhibited proliferation, while inducing differentiation, clear nuclear localisation of β-catenin, up-regulation of canonical Wnt target genes (Axin2, Lef1, Tcf4, Pitx2c and Lgr5) and activation of a TCF reporter construct. Again, concomitant knockdown of Axin1 and β-catenin in Axin2-null satellite cells rescued morphology and proliferation, but only partially prevented precocious differentiation. Thus, Axin1 and Axin2 do not have equivalent functions in satellite cells, but are both involved in repression of Wnt/β-catenin signalling to maintain proliferation and contribute to controlling timely myogenic differentiation.
Collapse
Affiliation(s)
- Nicolas Figeac
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, United Kingdom.
| | - Peter S Zammit
- King's College London, Randall Division of Cell & Molecular Biophysics, New Hunt's House, Guy's Campus, United Kingdom.
| |
Collapse
|
43
|
Brancaccio A, Palacios D. Chromatin signaling in muscle stem cells: interpreting the regenerative microenvironment. Front Aging Neurosci 2015; 7:36. [PMID: 25904863 PMCID: PMC4387924 DOI: 10.3389/fnagi.2015.00036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/04/2015] [Indexed: 12/12/2022] Open
Abstract
Muscle regeneration in the adult occurs in response to damage at expenses of a population of adult stem cells, the satellite cells. Upon injury, either physical or genetic, signals released within the satellite cell niche lead to the commitment, expansion and differentiation of the pool of muscle progenitors to repair damaged muscle. To achieve this goal satellite cells undergo a dramatic transcriptional reprogramming to coordinately activate and repress specific subset of genes. Although the epigenetics of muscle regeneration has been extensively discussed, less emphasis has been put on how extra-cellular cues are translated into the specific chromatin reorganization necessary for progression through the myogenic program. In this review we will focus on how satellite cells sense the regenerative microenvironment in physiological and pathological circumstances, paying particular attention to the mechanism through which the external stimuli are transduced to the nucleus to modulate chromatin structure and gene expression. We will discuss the pathways involved and how alterations in this chromatin signaling may contribute to satellite cells dysfunction during aging and disease.
Collapse
Affiliation(s)
- Arianna Brancaccio
- Laboratory of Epigenetics and Signaling, IRCCS Fondazione Santa Lucia Rome, Italy
| | - Daniela Palacios
- Laboratory of Epigenetics and Signaling, IRCCS Fondazione Santa Lucia Rome, Italy
| |
Collapse
|
44
|
WNT/β-Catenin Signaling Regulates Multiple Steps of Myogenesis by Regulating Step-Specific Targets. Mol Cell Biol 2015; 35:1763-76. [PMID: 25755281 DOI: 10.1128/mcb.01180-14] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 02/27/2015] [Indexed: 12/23/2022] Open
Abstract
Molecules involved in WNT/β-catenin signaling show specific spatiotemporal expression and play vital roles in myogenesis; however, it is still largely unknown how WNT/β-catenin signaling regulates each step of myogenesis. Here, we show that WNT/β-catenin signaling can control diverse biological processes of myogenesis by regulating step-specific molecules. In order to identify the temporally specific roles of WNT/β-catenin signaling molecules in muscle development and homeostasis, we used in vitro culture systems for both primary mouse myoblasts and C2C12 cells, which can differentiate into myofibers. We found that a blockade of WNT/β-catenin signaling in the proliferating cells decreases proliferation activity, but does not induce cell death, through the regulation of genes cyclin A2 (Ccna2) and cell division cycle 25C (Cdc25c). During muscle differentiation, the inhibition of WNT/β-catenin signaling blocks myoblast fusion through the inhibition of the Fermitin family homolog 2 (Fermt2) gene. Blocking WNT/β-catenin signaling in the well-differentiated myofibers results in the failure of maintenance of their structure by disruption of cadherin/β-catenin/actin complex formation, which plays a crucial role in connecting a myofiber's cytoskeleton to the surrounding extracellular matrix. Thus, our results indicate that WNT/β-catenin signaling can regulate multiple steps of myogenesis, including cell proliferation, myoblast fusion, and homeostasis, by targeting step-specific molecules.
Collapse
|
45
|
Suzuki A, Scruggs A, Iwata J. The temporal specific role of WNT/β-catenin signaling during myogenesis. JOURNAL OF NATURE AND SCIENCE 2015; 1:e143. [PMID: 26176019 PMCID: PMC4499510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Disruption of WNT/β-catenin signaling causes muscle developmental defects. However, it has been unclear how WNT/β-catenin signaling regulates each step of myogenesis. The in vitro culture of primary myoblasts and C2C12 cells (a myoblast cell line) has the ability to differentiate into myofibers in culture with differentiation inducers. These in vitro systems are useful to investigate each step of muscle development, ranging from cell proliferation to homeostasis, under the control of experimental conditions. Our recent study shows that WNT/β-catenin signaling can regulate myogenesis in a temporal specific manner by controlling the gene expression of cyclin A2 (Ccna2) and cell division cycle 25C (Cdc25c) during myoblast proliferation and fermitin family homolog 2 (Fermt2) during myoblast fusion and differentiation, respectively. In the well-differentiated myofibers, WNT/β-catenin signaling plays a role in the maintenance of their structure through a cadherin/β-catenin/actin complex formation, which is important for connecting a myofiber's cytoskeleton to the surrounding extracellular matrix. Thus, our recent study coupled with previous findings indicates that WNT/β-catenin signaling regulates myogenesis in a variety of ways, and any failure of these steps of myogenesis causes muscle developmental defects.
Collapse
Affiliation(s)
- Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, the University of Texas Health Science Center at Houston School of Dentistry, USA
- Center for Craniofacial Research, the University of Texas Health Science Center at Houston School of Dentistry, USA
| | - Anne Scruggs
- Department of Diagnostic & Biomedical Sciences, the University of Texas Health Science Center at Houston School of Dentistry, USA
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, the University of Texas Health Science Center at Houston School of Dentistry, USA
- Center for Craniofacial Research, the University of Texas Health Science Center at Houston School of Dentistry, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston, USA
| |
Collapse
|
46
|
Ma Z, Zhong Z, Zheng Z, Shi XM, Zhang W. Inhibition of glycogen synthase kinase-3β attenuates glucocorticoid-induced suppression of myogenic differentiation in vitro. PLoS One 2014; 9:e105528. [PMID: 25127359 PMCID: PMC4134315 DOI: 10.1371/journal.pone.0105528] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/24/2014] [Indexed: 11/18/2022] Open
Abstract
Glucocorticoids are the only therapy that has been demonstrated to alter the progress of Duchenne muscular dystrophy (DMD), the most common muscular dystrophy in children. However, glucocorticoids disturb skeletal muscle metabolism and hamper myogenesis and muscle regeneration. The mechanisms involved in the glucocorticoid-mediated suppression of myogenic differentiation are not fully understood. Glycogen synthase kinase-3β (GSK-3β) is considered to play a central role as a negative regulator in myogenic differentiation. Here, we showed that glucocorticoid treatment during the first 48 h in differentiation medium decreased the level of phosphorylated Ser9-GSK-3β, an inactive form of GSK-3β, suggesting that glucocorticoids affect GSK-3β activity. We then investigated whether GSK-3β inhibition could regulate glucocorticoid-mediated suppression of myogenic differentiation in vitro. Two methods were employed to inhibit GSK-3β: pharmacological inhibition with LiCl and GSK-3β gene knockdown. We found that both methods resulted in enhanced myotube formation and increased levels of muscle regulatory factors and muscle-specific protein expression. Importantly, GSK-3β inhibition attenuated glucocorticoid-induced suppression of myogenic differentiation. Collectively, these data suggest the involvement of GSK-3β in the glucocorticoid-mediated impairment of myogenic differentiation. Therefore, the inhibition of GSK-3β may be a strategy for preventing glucocorticoid-induced muscle degeneration.
Collapse
Affiliation(s)
- Zhenyu Ma
- Department of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhigang Zhong
- Department of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhenyang Zheng
- Department of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xing-Ming Shi
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia, United States of America
| | - Weixi Zhang
- Department of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- * E-mail:
| |
Collapse
|
47
|
Nagata Y, Ohashi K, Wada E, Yuasa Y, Shiozuka M, Nonomura Y, Matsuda R. Sphingosine-1-phosphate mediates epidermal growth factor-induced muscle satellite cell activation. Exp Cell Res 2014; 326:112-24. [DOI: 10.1016/j.yexcr.2014.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/25/2014] [Accepted: 06/16/2014] [Indexed: 01/03/2023]
|
48
|
Wu YJ, Fang YH, Chi HC, Chang LC, Chung SY, Huang WC, Wang XW, Lee KW, Chen SL. Insulin and LiCl synergistically rescue myogenic differentiation of FoxO1 over-expressed myoblasts. PLoS One 2014; 9:e88450. [PMID: 24551104 PMCID: PMC3923792 DOI: 10.1371/journal.pone.0088450] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 01/07/2014] [Indexed: 12/02/2022] Open
Abstract
Most recent studies reported that FoxO1 transcription factor was a negative regulator of myogenesis under serum withdrawal condition, a situation not actually found in vivo. Therefore, the role of FoxO1 in myogenesis should be re-examined under more physiologically relevant conditions. Here we found that FoxO1 was preferentially localized to nucleus in proliferating (PMB) and confluent myoblasts (CMB) and its nuclear exclusion was a prerequisite for formation of multinucleated myotubes (MT). The nuclear shuttling of FoxO1 in PMB could be prevented by leptomycin B and we further found that cytoplasmic accumulation of FoxO1 in myotubes was caused by the blockade of its nuclear import. Although over-expression of wildtype FoxO1 in C2C12 myoblasts significantly blocked their myogenic differentiation under serum withdrawal condition, application of insulin and LiCl, an activator of Wnt signaling pathway, to these cells successfully rescued their myogenic differentiation and generated myotubes with larger diameters. Interestingly, insulin treatment significantly reduced FoxO1 level and also delayed nuclear re-accumulation of FoxO1 triggered by mitogen deprivation. We further found that FoxO1 directly repressed the promoter activity of myogenic genes and this repression can be relieved by insulin and LiCl treatment. These results suggest that FoxO1 inhibits myogenesis in serum withdrawal condition but turns into a hypertrophy potentiator when other myogenic signals, such as Wnt and insulin, are available.
Collapse
Affiliation(s)
- Yi Ju Wu
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Yen Hsin Fang
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Hsiang Cheng Chi
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Li Chiung Chang
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Shih Ying Chung
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Wei Chieh Huang
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Xiao Wen Wang
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Kuan Wei Lee
- Department of Life Sciences, National Central University, Jhongli, Taiwan
| | - Shen Liang Chen
- Department of Life Sciences, National Central University, Jhongli, Taiwan
- * E-mail:
| |
Collapse
|
49
|
Fujimaki S, Hidaka R, Asashima M, Takemasa T, Kuwabara T. Wnt protein-mediated satellite cell conversion in adult and aged mice following voluntary wheel running. J Biol Chem 2014; 289:7399-412. [PMID: 24482229 DOI: 10.1074/jbc.m113.539247] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Muscle represents an abundant, accessible, and replenishable source of adult stem cells. Skeletal muscle-derived stem cells, called satellite cells, play essential roles in regeneration after muscle injury in adult skeletal muscle. Although the molecular mechanism of muscle regeneration process after an injury has been extensively investigated, the regulation of satellite cells under steady state during the adult stage, including the reaction to exercise stimuli, is relatively unknown. Here, we show that voluntary wheel running exercise, which is a low stress exercise, converts satellite cells to the activated state due to accelerated Wnt signaling. Our analysis showed that up-regulated canonical Wnt/β-catenin signaling directly modulated chromatin structures of both MyoD and Myf5 genes, resulting in increases in the mRNA expression of Myf5 and MyoD and the number of proliferative Pax7(+)Myf5(+) and Pax7(+) MyoD(+) cells in skeletal muscle. The effect of Wnt signaling on the activation of satellite cells, rather than Wnt-mediated fibrosis, was observed in both adult and aged mice. The association of β-catenin, T-cell factor, and lymphoid enhancer transcription factors of multiple T-cell factor/lymphoid enhancer factor regulatory elements, conserved in mouse, rat, and human species, with the promoters of both the Myf5 and MyoD genes drives the de novo myogenesis in satellite cells even in aged muscle. These results indicate that exercise-stimulated extracellular Wnts play a critical role in the regulation of satellite cells in adult and aged skeletal muscle.
Collapse
Affiliation(s)
- Shin Fujimaki
- From the Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-0046 and
| | | | | | | | | |
Collapse
|
50
|
Motohashi N, Asakura A. Muscle satellite cell heterogeneity and self-renewal. Front Cell Dev Biol 2014; 2:1. [PMID: 25364710 PMCID: PMC4206996 DOI: 10.3389/fcell.2014.00001] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/17/2023] Open
Abstract
Adult skeletal muscle possesses extraordinary regeneration capacities. After muscle injury or exercise, large numbers of newly formed muscle fibers are generated within a week as a result of expansion and differentiation of a self-renewing pool of muscle stem cells termed muscle satellite cells. Normally, satellite cells are mitotically quiescent and reside beneath the basal lamina of muscle fibers. Upon regeneration, satellite cells are activated, and give rise to daughter myogenic precursor cells. After several rounds of proliferation, these myogenic precursor cells contribute to the formation of new muscle fibers. During cell division, a minor population of myogenic precursor cells returns to quiescent satellite cells as a self-renewal process. Currently, accumulating evidence has revealed the essential roles of satellite cells in muscle regeneration and the regulatory mechanisms, while it still remains to be elucidated how satellite cell self-renewal is molecularly regulated and how satellite cells are important in aging and diseased muscle. The number of satellite cells is decreased due to the changing niche during ageing, resulting in attenuation of muscle regeneration capacity. Additionally, in Duchenne muscular dystrophy (DMD) patients, the loss of satellite cell regenerative capacity and decreased satellite cell number due to continuous needs for satellite cells lead to progressive muscle weakness with chronic degeneration. Thus, it is necessary to replenish muscle satellite cells continuously. This review outlines recent findings regarding satellite cell heterogeneity, asymmetric division and molecular mechanisms in satellite cell self-renewal which is crucial for maintenance of satellite cells as a muscle stem cell pool throughout life. In addition, we discuss roles in the stem cell niche for satellite cell maintenance, as well as related cell therapies for approaching treatment of DMD.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Neurology, Paul and Sheila Wellstone Muscular Dystrophy Center, Stem Cell Institute, University of Minnesota Medical School Minneapolis, MN, USA
| | - Atsushi Asakura
- Department of Neurology, Paul and Sheila Wellstone Muscular Dystrophy Center, Stem Cell Institute, University of Minnesota Medical School Minneapolis, MN, USA
| |
Collapse
|