Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 932
1.
Curr Top Dev Biol ; 158: 53-82, 2024.
Article En | MEDLINE | ID: mdl-38670716

Myocyte fusion is a pivotal process in the development and regeneration of skeletal muscle. Failure during fusion can lead to a range of developmental as well as pathological consequences. This review aims to comprehensively explore the intricate processes underlying myocyte fusion, from the molecular to tissue scale. We shed light on key players, such as the muscle-specific fusogens - Myomaker and Myomixer, in addition to some lesser studied molecules contributing to myocyte fusion. Conserved across vertebrates, Myomaker and Myomixer play a crucial role in driving the merger of plasma membranes of fusing myocytes, ensuring the formation of functional muscle syncytia. Our multiscale approach also delves into broader cell and tissue dynamics that orchestrate the timing and positioning of fusion events. In addition, we explore the relevance of muscle fusogens to human health and disease. Mutations in fusogen genes have been linked to congenital myopathies, providing unique insights into the molecular basis of muscle diseases. We conclude with a discussion on potential therapeutic avenues that may emerge from manipulating the myocyte fusion process to remediate skeletal muscle disorders.


Cell Fusion , Humans , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle Cells/metabolism , Muscle Cells/cytology , Muscle Proteins/metabolism , Muscle Proteins/genetics
2.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article En | MEDLINE | ID: mdl-35163664

Cardiac patch implantation helps maximize the paracrine function of grafted cells and serves as a reservoir of soluble proangiogenic factors required for the neovascularization of infarcted hearts. We have previously fabricated a cardiac patch, EF-HAM, composed of a human amniotic membrane (HAM) coated with aligned PLGA electrospun fibers (EF). In this study, we aimed to evaluate the biocompatibility and angiogenic effects of EF-HAM scaffolds with varying fiber thicknesses on the paracrine behavior of skeletal muscle cells (SkM). Conditioned media (CM) obtained from SkM-seeded HAM and EF-HAM scaffolds were subjected to multiplex analysis of angiogenic factors and tested on HUVECs for endothelial cell viability, migration, and tube formation analyses. All three different groups of EF-HAM scaffolds demonstrated excellent biocompatibility with SkM. CM derived from SkM-seeded EF-HAM 7 min scaffolds contained significantly elevated levels of proangiogenic factors, including angiopoietin-1, IL-8, and VEGF-C compared to plain CM, which was obtained from SkM cultured on the plain surface. CM obtained from all SkM-seeded EF-HAM scaffolds significantly increased the viability of HUVECs compared to plain CM after five days of culture. However, only EF-HAM 7 min CM induced a higher migration capacity in HUVECs and formed a longer and more elaborate capillary-like network on Matrigel compared with plain CM. Surface roughness and wettability of EF-HAM 7 min scaffolds might have influenced the proportion of skeletal myoblasts and fibroblasts growing on the scaffolds and subsequently potentiated the angiogenic paracrine function of SkM. This study demonstrated the angioinductive properties of EF-HAM composite scaffold and its potential applications in the repair and regeneration of ischemic tissues.


Ischemia/therapy , Neovascularization, Physiologic , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Regeneration/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Amnion , Angiopoietin-1/metabolism , Biocompatible Materials/chemistry , Cell Movement , Cell Survival , Culture Media, Conditioned/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-8/metabolism , Ischemia/pathology , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Cells/ultrastructure , Muscle, Skeletal/cytology , Vascular Endothelial Growth Factor A/metabolism
3.
Endocrinology ; 163(1)2022 01 01.
Article En | MEDLINE | ID: mdl-34534278

Cross-talk between peripheral tissues is essential to ensure the coordination of nutrient intake with disposition during the feeding period, thereby preventing metabolic disease. This mini-review considers the interactions between the key peripheral tissues that constitute the metabolic clock, each of which is considered in a separate mini-review in this collation of articles published in Endocrinology in 2020 and 2021, by Martchenko et al (Circadian rhythms and the gastrointestinal tract: relationship to metabolism and gut hormones); Alvarez et al (The microbiome as a circadian coordinator of metabolism); Seshadri and Doucette (Circadian regulation of the pancreatic beta cell); McCommis et al (The importance of keeping time in the liver); Oosterman et al (The circadian clock, shift work, and tissue-specific insulin resistance); and Heyde et al (Contributions of white and brown adipose tissues to the circadian regulation of energy metabolism). The use of positive- and negative-feedback signals, both hormonal and metabolic, between these tissues ensures that peripheral metabolic pathways are synchronized with the timing of food intake, thus optimizing nutrient disposition and preventing metabolic disease. Collectively, these articles highlight the critical role played by the circadian clock in maintaining metabolic homeostasis.


Circadian Clocks/physiology , Circadian Rhythm , Feeding Behavior , Homeostasis , Liver/physiology , Adipocytes/cytology , Animals , Endocrinology/methods , Energy Intake , Energy Metabolism/physiology , Feedback, Physiological , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Intestines/physiology , Islets of Langerhans/cytology , Mammals/physiology , Metabolic Diseases/metabolism , Microbiota , Models, Biological , Muscle Cells/cytology , Muscle, Skeletal/physiology
4.
Dev Biol ; 483: 1-12, 2022 03.
Article En | MEDLINE | ID: mdl-34963554

The ascidian larval tail contains muscle cells for swimming. Most of these muscle cells differentiate autonomously. The genetic program behind this autonomy has been studied extensively and the genetic cascade from maternal factors to initiation of expression of a muscle structural gene, Myl.c, has been uncovered; Myl.c expression is directed initially by transcription factor Tbx6-r.b at the 64-cell stage and then by the combined actions of Tbx6-r.b and Mrf from the gastrula to early tailbud stages. In the present study, we showed that transcription of Myl.c continued in late tailbud embryos and larvae, although a fusion protein of Tbx6-r.b and GFP was hardly detectable in late tailbud embryos. A knockdown experiment, reporter assay, and in vitro binding assay indicated that an essential cis-regulatory element of Myl.c that bound Tbx6-r.b in early embryos bound Tbx15/18/22 in late embryos to maintain expression of Myl.c. We also found that Tbx15/18/22 was controlled by Mrf, which constitutes a regulatory loop with Tbx6-r.b. Therefore, our data indicated that Tbx15/18/22 was activated initially under control of this regulatory loop as in the case of Myl.c, and then Tbx15/18/22 maintained the expression of Myl.c after Tbx6-r.b had disappeared. RNA-sequencing of Tbx15/18/22 morphant embryos revealed that many muscle structural genes were regulated similarly by Tbx15/18/22. Thus, the present study revealed the mechanisms of maintenance of transcription of muscle structural genes in late embryos in which Tbx15/18/22 takes the place of Tbx6-r.b.


Gene Expression Regulation, Developmental , Gene Expression , Muscles/embryology , Muscles/metabolism , T-Box Domain Proteins/metabolism , Urochordata/embryology , Urochordata/genetics , Animals , Binding Sites , Cell Differentiation/genetics , Female , Gastrula/metabolism , Gene Knockdown Techniques , Gene Regulatory Networks , Muscle Cells/cytology , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Oviparity/genetics , T-Box Domain Proteins/genetics , Transcription, Genetic/genetics
5.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article En | MEDLINE | ID: mdl-34830317

Clostridium sporogenes (C. sporogenes), as a potential probiotic, metabolizes tryptophan and produces an anti-inflammatory metabolite, indole-3-propionic acid (IPA). Herein, we studied the effects of C. sporogenes and its bioactive metabolite, IPA, on skeletal muscle development and chronic inflammation in mice. In the in vivo study, the muscle tissues and serum samples of mice with C. sporogenes supplementation were used to analyze the effects of C. sporogenes on muscle metabolism; the IPA content was determined by metabonomics and ELISA. In an in vitro study, C2C12 cells were exposed to lipopolysaccharide (LPS) alone or LPS + IPA to verify the effect of IPA on muscle cell inflammation by transcriptome, and the involved mechanism was revealed by different functional assays. We observed that C. sporogenes colonization significantly increased the body weight and muscle weight gain, as well as the myogenic regulatory factors' (MRFs) expression. In addition, C. sporogenes significantly improved host IPA content and decreased pro-inflammatory cytokine levels in the muscle tissue of mice. Subsequently, we confirmed that IPA promoted C2C12 cells' proliferation by activating MRF signaling. IPA also effectively protected against LPS-induced C2C12 cells inflammation by activating Pregnane X Receptor and restoring the inhibited miR-26a-2-3p expression. miR-26a-2-3p serves as a novel muscle inflammation regulatory factor that could directly bind to the 3'-UTR of IL-1ß, a key initiator factor in inflammation. The results suggested that C. sporogenes with its functional metabolite IPA not only helps muscle growth development, but also protects against inflammation, partly by the IPA/ miR-26a-2-3p /IL-1ß cascade.


Clostridium/metabolism , Indoles/metabolism , Interleukin-1beta/genetics , MicroRNAs/genetics , Muscle Cells/drug effects , Pregnane X Receptor/genetics , Propionates/metabolism , 3' Untranslated Regions , Animals , Cell Line , Gastrointestinal Microbiome/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental , Indoles/pharmacology , Inflammation/prevention & control , Interleukin-1beta/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Pregnane X Receptor/metabolism , Probiotics/metabolism , Propionates/pharmacology , Signal Transduction , Transcriptome , Tryptophan/metabolism
6.
Elife ; 102021 10 04.
Article En | MEDLINE | ID: mdl-34605406

Non-centrosomal microtubule-organizing centers (MTOCs) are pivotal for the function of multiple cell types, but the processes initiating their formation are unknown. Here, we find that the transcription factor myogenin is required in murine myoblasts for the localization of MTOC proteins to the nuclear envelope. Moreover, myogenin is sufficient in fibroblasts for nuclear envelope MTOC (NE-MTOC) formation and centrosome attenuation. Bioinformatics combined with loss- and gain-of-function experiments identified induction of AKAP6 expression as one central mechanism for myogenin-mediated NE-MTOC formation. Promoter studies indicate that myogenin preferentially induces the transcription of muscle- and NE-MTOC-specific isoforms of Akap6 and Syne1, which encodes nesprin-1α, the NE-MTOC anchor protein in muscle cells. Overexpression of AKAP6ß and nesprin-1α was sufficient to recruit endogenous MTOC proteins to the nuclear envelope of myoblasts in the absence of myogenin. Taken together, our results illuminate how mammals transcriptionally control the switch from a centrosomal MTOC to an NE-MTOC and identify AKAP6 as a novel NE-MTOC component in muscle cells.


A Kinase Anchor Proteins/metabolism , Microtubule-Organizing Center/physiology , Muscle Cells/metabolism , Myogenin/metabolism , 3T3 Cells , Animals , Cell Line , HEK293 Cells , Humans , Mice , Muscle Cells/cytology , Nuclear Envelope
7.
Nat Genet ; 53(10): 1480-1492, 2021 10.
Article En | MEDLINE | ID: mdl-34611363

Higher-order chromatin structure regulates gene expression, and mutations in proteins mediating genome folding underlie developmental disorders known as cohesinopathies. However, the relationship between three-dimensional genome organization and embryonic development remains unclear. Here we define a role for bromodomain-containing protein 4 (BRD4) in genome folding, and leverage it to understand the importance of genome folding in neural crest progenitor differentiation. Brd4 deletion in neural crest results in cohesinopathy-like phenotypes. BRD4 interacts with NIPBL, a cohesin agonist, and BRD4 depletion or loss of the BRD4-NIPBL interaction reduces NIPBL occupancy, suggesting that BRD4 stabilizes NIPBL on chromatin. Chromatin interaction mapping and imaging experiments demonstrate that BRD4 depletion results in compromised genome folding and loop extrusion. Finally, mutation of individual BRD4 amino acids that mediate an interaction with NIPBL impedes neural crest differentiation into smooth muscle. Remarkably, loss of WAPL, a cohesin antagonist, rescues attenuated smooth muscle differentiation resulting from BRD4 loss. Collectively, our data reveal that BRD4 choreographs genome folding and illustrates the relevance of balancing cohesin activity for progenitor differentiation.


Cell Differentiation , Genome , Neural Crest/cytology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Integrases/metabolism , Mice , Models, Biological , Mouse Embryonic Stem Cells/metabolism , Muscle Cells/cytology , Neural Crest/metabolism , Protein Binding , Protein Domains , Proteolysis , Transcription Factors/chemistry , Transcription, Genetic , Cohesins
8.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article En | MEDLINE | ID: mdl-34638875

Pluripotent adult stem cells have potential applications in cell therapy and tissue engineering. Urine-derived stem cells (UDSCs) differentiate into various cell types. Here, we attempted to differentiate human UDSCs (hUDSCs) into smooth muscle cells (SMCs) using transforming growth factor-beta 1 (TGF-ß1) and/or PD98059, an extracellular signal-regulated kinase (ERK) inhibitor. Both quantitative polymerase chain reaction (qPCR) and Western blot analysis showed that the expression of messenger ribonucleic acid (mRNA) and proteins for alpha-smooth muscle actin (α-SMA), calponin (CNN1), and smooth muscle myosin heavy chain (SM-MHC), which are specific markers for SMCs, increased on day 9 after differentiation and again on day 14. The differentiated cells from human UDSCs (hUDSCs) with a combination of TGF-ß1 and PD98059 showed the highest expression of SMC marker proteins. Immunocytochemical staining performed to assess the molecular expression revealed CNN and α-SMA colocalizing in the cytoplasm. The cells that differentiated from hUDSCs with a combination of TGF-ß1 and PD98059 showed the strongest expression for CNN1, α-SMA, and SM-MHC. Functional testing of the differentiated cells revealed a stronger contractile capacity for the cells differentiated with a combination of PD98059 and TGF-ß1 than those differentiated with a single factor. These results suggest the combination of PD98059 and TGF-ß1 to be a more effective differentiation method and that differentiated SMCs could be used for restoring the functions of the sphincter muscle or bladder.


Cell Differentiation/drug effects , Flavonoids/pharmacology , Muscle Cells , Stem Cells , Transforming Growth Factor beta1/pharmacology , Urine/cytology , Adult , Female , Humans , Male , Middle Aged , Muscle Cells/cytology , Muscle Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism
9.
Int J Mol Sci ; 22(17)2021 Sep 06.
Article En | MEDLINE | ID: mdl-34502539

Muscular dystrophies are a heterogeneous group of inherited diseases characterized by the progressive degeneration and weakness of skeletal muscles, leading to disability and, often, premature death. To date, no effective therapies are available to halt or reverse the pathogenic process, and meaningful treatments are urgently needed. From this perspective, it is particularly important to establish reliable in vitro models of human muscle that allow the recapitulation of disease features as well as the screening of genetic and pharmacological therapies. We herein review and discuss advances in the development of in vitro muscle models obtained from human induced pluripotent stem cells, which appear to be capable of reproducing the lack of myofiber proteins as well as other specific pathological hallmarks, such as inflammation, fibrosis, and reduced muscle regenerative potential. In addition, these platforms have been used to assess genetic correction strategies such as gene silencing, gene transfer and genome editing with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), as well as to evaluate novel small molecules aimed at ameliorating muscle degeneration. Furthermore, we discuss the challenges related to in vitro drug testing and provide a critical view of potential therapeutic developments to foster the future clinical translation of preclinical muscular dystrophy studies.


Cell Differentiation/physiology , Drug Discovery/methods , Genetic Therapy/methods , Induced Pluripotent Stem Cells/physiology , Muscle Cells/physiology , Muscular Dystrophies/therapy , Animals , Dystrophin/genetics , Dystrophin/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Muscle Cells/cytology , Muscular Dystrophies/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy
10.
Int J Mol Sci ; 22(18)2021 Sep 10.
Article En | MEDLINE | ID: mdl-34575979

MicroRNAs (miRNAs) play an essential role in the regulation of a number of physiological functions. miR-133a and other muscular miRs (myomiRs) play a key role in muscle cell growth and in some type of cancers. Here, we show that miR133a is upregulated in individuals that undertake physical exercise. We used a skeletal muscle differentiation model to dissect miR-133a's role and to identify new targets, identifying Tropomyosin-4 (TPM4). This protein is expressed during muscle differentiation, but importantly it is an essential component of microfilament cytoskeleton and stress fibres formation. The microfilament scaffold remodelling is an essential step in cell transformation and tumour progression. Using the muscle system, we obtained valuable information about the microfilament proteins, and the knowledge on these molecular players can be transferred to the cytoskeleton rearrangement observed in cancer cells. Further investigations showed a role of TPM4 in cancer physiology, specifically, we found that miR-133a downregulation leads to TPM4 upregulation in colon carcinoma (CRC), and this correlates with a lower patient survival. At molecular level, we demonstrated in myocyte differentiation that TPM4 is positively regulated by the TA isoform of the p63 transcription factor. In muscles, miR-133a generates a myogenic stimulus, reducing the differentiation by downregulating TPM4. In this system, miR-133a counteracts the differentiative TAp63 activity. Interestingly, in CRC cell lines and in patient biopsies, miR-133a is able to regulate TPM4 activity, while TAp63 is not active. The downregulation of the miR leads to TPM4 overexpression, this modifies the architecture of the cell cytoskeleton contributing to increase the invasiveness of the tumour and associating with a poor prognosis. These results add data to the interesting question about the link between physical activity, muscle physiology and protection against colorectal cancer. The two phenomena have in common the cytoskeleton remodelling, due to the TPM4 activity, that is involved in stress fibres formation.


Cell Differentiation/genetics , Colonic Neoplasms/genetics , MicroRNAs/genetics , Transcription Factors/genetics , Tropomyosin/genetics , Tumor Suppressor Proteins/genetics , Actin Cytoskeleton/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/pathology , Cytoskeleton/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Muscle Cells/cytology , Muscle Development/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Stress Fibers/genetics
11.
Methods Mol Biol ; 2319: 137-141, 2021.
Article En | MEDLINE | ID: mdl-34331251

Lymphatic muscle cells (LMCs), with unique characteristics resembling a combination of both cardiac and smooth muscle cells, play an essential role in the spontaneous contraction of the lymphatic vessels to pump fluid forward. However, our understanding of the more detailed molecular phenotypes of LMCs is limited. Here, we described a method to isolate the LMCs from rat mesentery and then culture the cells in vitro, which will serve a lot more molecular biology study of LMCs and significantly improve our knowledge about the unique characteristics of LMCs.


Cell Culture Techniques/methods , Cell Separation/methods , Dissection/methods , Mesentery/cytology , Muscle Cells/cytology , Animals , Fluorescent Antibody Technique , Muscle Cells/metabolism , Rats
12.
Molecules ; 26(9)2021 May 03.
Article En | MEDLINE | ID: mdl-34063700

Momordica charantia is a popular vegetable associated with effective complementary and alternative diabetes management in some parts of the world. However, the molecular mechanism is less commonly investigated. In this study, we investigated the association between a major cucurbitane triterpenoid isolated from M. charantia, 3ß,7ß,25-trihydroxycucurbita-5,23(E)-dien-19-al (THCB) and peroxisome proliferator activated receptor gamma (PPARγ) activation and its related activities using cell culture and molecular biology techniques. In this study, we report on both M. charantia fruit crude extract and THCB in driving the luciferase activity of Peroxisome Proliferator Response Element, associated with PPARγ activation. Other than that, THCB also induced adipocyte differentiation at far less intensity as compared to the full agonist rosiglitazone. In conjunction, THCB treatment on adipocytes also resulted in upregulation of PPAR gamma target genes expression; AP2, adiponectin, LPL and CD34 at a lower magnitude compared to rosiglitazone's induction. THCB also induced glucose uptake into muscle cells and the mechanism is via Glut4 translocation to the cell membrane. In conclusion, THCB acts as one of the many components in M. charantia to induce hypoglycaemic effect by acting as PPARγ ligand and inducing glucose uptake activity in the muscles by means of Glut4 translocation.


Momordica/chemistry , PPAR gamma/metabolism , Triterpenes/chemistry , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Differentiation , Cell Membrane/metabolism , Glucose/metabolism , Hepatocytes/cytology , Hypoglycemia/drug therapy , Insulin/chemistry , Ligands , Mice , Muscle Cells/cytology , Protein Domains , Rosiglitazone/pharmacology , Triterpenes/pharmacology
13.
Growth Horm IGF Res ; 59: 101406, 2021 08.
Article En | MEDLINE | ID: mdl-34126555

OBJECTIVE: IGF-I and branched-chain amino acids have been reported to promote muscle hypertrophy via the stimulation of protein synthesis. Sestrin2, the function of which is regulated by leucine, has been reported to attenuate the activity of the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) that stimulates protein synthesis. The objective of this study was to examine whether IGF-I modulates Sestrin2 abundance and to clarify the involvement of Sestrin2 in the effect of IGF-I and leucine on mTROC1. DESIGN: C2C12 and L6 myocytes were stimulated by leucine (1 mM) with or without pretreatment with IGF-I (100 ng/mL). Phosphorylation of p70 S6 kinase (S6K) and 4E-binding protein 1 (4E-BP1), both of which are targets of the mTORC1, was examined by western blotting. Effects of Sestrin2 small interfering RNA (siRNA) on the actions of leucine and IGF-I were examined. Sestrin2 mRNA and protein levels were also determined after Sestrin2 siRNA. RESULTS: Leucine increased the phosphorylation of S6K and 4E-BP1 in a dose-dependent manner. Pretreatment with IGF-I for 5 h further increased the stimulatory effect of leucine on the phosphorylation of S6K and 4E-BP1 in C2C12 cells. IGF-I increased Sestrin2 protein and messenger RNA levels. Sestrin2 siRNA increased or tended to increase basal phosphorylation of 4E-BP1 and decreased the leucine-induced phosphorylation in C2C12 and L6 cells, in particular after IGF-I treatment, suggesting the involvement of Sestrin2 in the action of leucine and IGF-I. The net increase in leucine-induced 4E-BP1 phosphorylation appeared to be attenuated by Sestrin2 siRNA. Likewise, Sestrin2 siRNA attenuated leucine-induced S6K phosphorylation in L6 cells. However, Sestrin2 siRNA did not influence leucine-induced S6K phosphorylation in C2C12 cells. CONCLUSIONS: IGF-I and leucine cooperatively increased mTORC1 activity in C2C12 cells. IGF-I increased Sestrin2. Sestrin2 siRNA experiments showed that Sestrin2 was involved in the effect of leucine and IGF-I on mTORC1 activity in C2C12 and L6 cells, and suggested that increased Sestrin2 by IGF-I pretreatment might play a role in enhancing the effect of leucine on mTORC1.


Gene Expression Regulation/drug effects , Insulin-Like Growth Factor I/pharmacology , Leucine/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Muscle Cells/metabolism , Peroxidases/metabolism , Animals , Cells, Cultured , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Muscle Cells/cytology , Muscle Cells/drug effects , Peroxidases/genetics , Phosphorylation , Signal Transduction
14.
Genes Cells ; 26(7): 495-512, 2021 Jul.
Article En | MEDLINE | ID: mdl-33960573

Mesenchymal stem cells (MSCs) are the likely precursors of multiple lines of mesenchymal cells. The existence of bona fide MSCs with self-renewal capacity and differentiation potential into all mesenchymal lineages, however, has been unclear because of the lack of MSC-specific marker(s) that are not expressed by the terminally differentiated progeny. Meflin, a glycosylphosphatidylinositol-anchored protein, is an MSC marker candidate that is specifically expressed in rare stromal cells in all tissues. Our previous report showed that Meflin expression becomes down-regulated in bone marrow-derived MSCs cultured on plastic, making it difficult to examine the self-renewal and differentiation of Meflin-positive cells at the single-cell level. Here, we traced the lineage of Meflin-positive cells in postnatal and adult mice, showing that those cells differentiated into white and brown adipocytes, osteocytes, chondrocytes and skeletal myocytes. Interestingly, cells derived from Meflin-positive cells formed clusters of differentiated cells, implying the in situ proliferation of Meflin-positive cells or their lineage-committed progenitors. These results, taken together with previous findings that Meflin expression in cultured MSCs was lost upon their multilineage differentiation, suggest that Meflin is a useful potential marker to localize MSCs and/or their immature progenitors in multiple tissues.


Cell Differentiation , Cell Lineage , Immunoglobulins/metabolism , Mesenchymal Stem Cells/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Chondrocytes/cytology , Chondrocytes/metabolism , Immunoglobulins/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Muscle Cells/cytology , Muscle Cells/metabolism , Osteocytes/cytology , Osteocytes/metabolism
15.
PLoS One ; 16(5): e0250741, 2021.
Article En | MEDLINE | ID: mdl-33961664

The interactions between skeletal muscle and bone have been recently noted, and muscle-derived humoral factors related to bone metabolism play crucial roles in the muscle/bone relationships. We previously reported that extracellular vesicles from mouse muscle C2C12 cells (Myo-EVs) suppress osteoclast formation in mice. Although mechanical stress is included in extrinsic factors which are important for both muscle and bone, the detailed roles of mechanical stress in the muscle/bone interactions have still remained unknown. In present study, we examined the effects of fluid flow shear stress (FFSS) to C2C12 cells on the physiological actions of muscle cell-derived EV. Applying FFSS to C2C12 cells significantly enhanced muscle cell-derived EV-suppressed osteoclast formation and several osteoclast-related gene levels in mouse bone marrow cells in the presence of receptor activator nuclear factor κB ligand (RANKL). Moreover, FFSS to C2C12 cells significantly enhanced muscle cell-derived EV-suppressed mitochondria biogenesis genes during osteoclast formation with RANKL treatment. In addition, FFSS to C2C12 cells significantly enhanced muscle cell-derived EV-suppressed osteoclast formation and several osteoclast-related gene levels in Raw264.7 cells in the presence of RANKL. Small RNA-seq-analysis showed that FFSS elevated the expression of miR196a-5p and miR155-5p with the suppressive actions of osteoclast formation and low expression in mouse bone cells. On the other hand, muscle cell-derived EVs with or without FFSS to C2C12 cells did not affect the expression of osteogenic genes, alkaline phosphatase activity and mineralization in mouse osteoblasts. In conclusion, we first showed that FFSS to C2C12 cells enhances the suppressive effects of muscle cell-derived EVs on osteoclast formation in mouse cells. Muscle cell-derived EVs might be partly involved in the effects of mechanical stress on the muscle/bone relationships.


Bone and Bones/physiology , Extracellular Vesicles/metabolism , Hydrodynamics , Muscle Cells/cytology , Shear Strength , Stress, Mechanical , Animals , Biomechanical Phenomena , Cell Line , Mice
16.
J Vet Med Sci ; 83(7): 1022-1030, 2021 Jul 02.
Article En | MEDLINE | ID: mdl-33967186

This study aimed to investigate the function of estrogen receptors (ERs) in myoregeneration and intermuscular adipogenesis. Ovariectomized (OVX) ERα knockout (KO) mice and ERß KO mice were used to assess the effect of estrogen on the myoregenerative process. Tibialis anterior muscle was collected on days 7, 10, and 14 after cardiotoxin injection to assess myotube morphology and adipogenesis area. Regenerated myotubes from OVX-ERß KO mice were consistently smaller in diameter than those from OVX-ERα KO and OVX-wild-type mice, whereas the adipogenesis area of OVX-ERß KO mice was consistently greater than that of the other types. Therefore, ERß may be an influential factor in promoting myoregeneration and adipogenesis inhibition compared to ERα.


Adipogenesis , Estrogen Receptor alpha , Estrogen Receptor beta , Muscle Cells/cytology , Regeneration , Animals , Estradiol , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogens , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovariectomy/veterinary
17.
Cells ; 10(4)2021 03 27.
Article En | MEDLINE | ID: mdl-33801626

Muscle tissue is often removed during hamstring tendon graft preparation for anterior cruciate ligament (ACL) reconstruction. The purpose of the study was to test whether preservation of muscle remnants on a tendon graft is beneficial to the graft healing process following ACL reconstruction. Co-culturing of tendon-derived cells (TDCs) and muscle-derived cells (MDCs) was performed at various ratios, and their potential for cell viability and multilineage differentiation was compared to a single TDC cell group. Ligamentous and chondrogenic differentiation was most enhanced when a small population of MDCs was co-cultured with TDCs (6:2 co-culture group). Cell viability and osteogenic differentiation were proportionally enhanced with increasing MDC population size. MDCs co-cultured with TDCs possess both the ability to enhance cell viability and differentiate into other cell lineages.


Cell Differentiation , Hamstring Tendons/transplantation , Muscle Cells/cytology , Preservation, Biological , Adolescent , Adult , Becaplermin/pharmacology , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Coculture Techniques , Collagen/biosynthesis , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Ligaments/cytology , Male , Muscle Cells/drug effects , Osteogenesis/drug effects , Young Adult
18.
Int J Mol Sci ; 22(5)2021 Mar 01.
Article En | MEDLINE | ID: mdl-33804428

The mammalian ventricular myocardium forms a functional syncytium due to flow of electrical current mediated in part by gap junctions localized within intercalated disks. The connexin (Cx) subunit of gap junctions have direct and indirect roles in conduction of electrical impulse from the cardiac pacemaker via the cardiac conduction system (CCS) to working myocytes. Cx43 is the dominant isoform in these channels. We have studied the distribution of Cx43 junctions between the CCS and working myocytes in a transgenic mouse model, which had the His-Purkinje portion of the CCS labeled with green fluorescence protein. The highest number of such connections was found in a region about one-third of ventricular length above the apex, and it correlated with the peak proportion of Purkinje fibers (PFs) to the ventricular myocardium. At this location, on the septal surface of the left ventricle, the insulated left bundle branch split into the uninsulated network of PFs that continued to the free wall anteriorly and posteriorly. The second peak of PF abundance was present in the ventricular apex. Epicardial activation maps correspondingly placed the site of the first activation in the apical region, while some hearts presented more highly located breakthrough sites. Taken together, these results increase our understanding of the physiological pattern of ventricular activation and its morphological underpinning through detailed CCS anatomy and distribution of its gap junctional coupling to the working myocardium.


Cell Communication , Connexin 43/physiology , Gap Junctions/physiology , Heart Ventricles/pathology , Muscle Cells/physiology , Pericardium/physiology , Purkinje Fibers/physiology , Animals , Female , Male , Mice , Muscle Cells/cytology , Pericardium/cytology , Purkinje Fibers/cytology
19.
Biosci Biotechnol Biochem ; 85(5): 1227-1234, 2021 Apr 24.
Article En | MEDLINE | ID: mdl-33704409

Among many factors of controlling stem cell differentiation, the key transcription factor upregulation via physical force is a good strategy on the lineage-specific differentiation of stem cells. The study aimed to compare growth and myogenic potentials between the parental cells (PCs) and the 1-day-old C2C12 spheroid-derived cells (SDCs) in two-dimensional (2D) and three-dimensional (3D) culture conditions through examination of the cell proliferation and the expression of myogenic genes. The data showed that 1-day-old spheroids had more intense expression of MyoD gene with respect to the PCs. The proliferation of the SDCs is significantly higher than the PCs in a time-dependent manner. The SDCs had also significantly higher myogenic potential than the PCs in 2D and 3D culture conditions. The results suggest that MyoD gene upregulation through cell-cell contacts is the good approach for preparation of seed cells in muscle tissue engineering.


Cell Culture Techniques , Muscle Cells/metabolism , Muscle Development/genetics , MyoD Protein/genetics , Myoblasts/metabolism , Spheroids, Cellular/metabolism , Actinin/genetics , Actinin/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Collagen/chemistry , Collagen/pharmacology , Gene Expression Regulation , Mice , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Development/drug effects , MyoD Protein/antagonists & inhibitors , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myogenin/genetics , Myogenin/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Tissue Engineering/methods
20.
J Biol Chem ; 296: 100606, 2021.
Article En | MEDLINE | ID: mdl-33789162

In addition to maintaining cellular ER Ca2+ stores, store-operated Ca2+ entry (SOCE) regulates several Ca2+-sensitive cellular enzymes, including certain adenylyl cyclases (ADCYs), enzymes that synthesize the secondary messenger cyclic AMP (cAMP). Ca2+, acting with calmodulin, can also increase the activity of PDE1-family phosphodiesterases (PDEs), which cleave the phosphodiester bond of cAMP. Surprisingly, SOCE-regulated cAMP signaling has not been studied in cells expressing both Ca2+-sensitive enzymes. Here, we report that depletion of ER Ca2+ activates PDE1C in human arterial smooth muscle cells (HASMCs). Inhibiting the activation of PDE1C reduced the magnitude of both SOCE and subsequent Ca2+/calmodulin-mediated activation of ADCY8 in these cells. Because inhibiting or silencing Ca2+-insensitive PDEs had no such effects, these data identify PDE1C-mediated hydrolysis of cAMP as a novel and important link between SOCE and its activation of ADCY8. Functionally, we showed that PDE1C regulated the formation of leading-edge protrusions in HASMCs, a critical early event in cell migration. Indeed, we found that PDE1C populated the tips of newly forming leading-edge protrusions in polarized HASMCs, and co-localized with ADCY8, the Ca2+ release activated Ca2+ channel subunit, Orai1, the cAMP-effector, protein kinase A, and an A-kinase anchoring protein, AKAP79. Because this polarization could allow PDE1C to control cAMP signaling in a hyper-localized manner, we suggest that PDE1C-selective therapeutic agents could offer increased spatial specificity in HASMCs over agents that regulate cAMP globally in cells. Similarly, such agents could also prove useful in regulating crosstalk between Ca2+/cAMP signaling in other cells in which dysregulated migration contributes to human pathology, including certain cancers.


Arteries/cytology , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Muscle Cells/cytology , Signal Transduction , Biological Transport , Cell Movement , Gene Expression Regulation, Enzymologic , Humans , Kinetics
...