Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Proc Natl Acad Sci U S A ; 121(21): e2317495121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38753506

ABSTRACT

Myogenic regeneration relies on the proliferation and differentiation of satellite cells. TECRL (trans-2,3-enoyl-CoA reductase like) is an endoplasmic reticulum protein only expressed in cardiac and skeletal muscle. However, its role in myogenesis remains unknown. We show that TECRL expression is increased in response to injury. Satellite cell-specific deletion of TECRL enhances muscle repair by increasing the expression of EGR2 through the activation of the ERK1/2 signaling pathway, which in turn promotes the expression of PAX7. We further show that TECRL deletion led to the upregulation of the histone acetyltransferase general control nonderepressible 5, which enhances the transcription of EGR2 through acetylation. Importantly, we showed that AAV9-mediated TECRL silencing improved muscle repair in mice. These findings shed light on myogenic regeneration and muscle repair.


Subject(s)
Early Growth Response Protein 2 , Muscle Development , Muscle, Skeletal , Regeneration , Animals , Mice , Muscle, Skeletal/metabolism , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/genetics , Muscle Development/genetics , Regeneration/genetics , Up-Regulation , Satellite Cells, Skeletal Muscle/metabolism , PAX7 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , MAP Kinase Signaling System , Mice, Knockout , Cell Differentiation
2.
Cell Mol Life Sci ; 77(5): 937-952, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31312880

ABSTRACT

BACKGROUND AND AIMS: Allogeneic human umbilical mesenchymal stem cells (alloUMSC) are convenient cell source for stem cell-based therapy. However, immune rejection is a major obstacle for clinical application of alloUMSC for cardiac repair after myocardial infarction (MI). The immune rejection is due to the presence of human leukocyte antigen (HLA) class I molecule which is increased during MI. The aim of this study was to knockout HLA light chain ß2-microglobulin (B2M) in UMSC to enhance stem cell engraftment and survival after transplantation. METHODS AND RESULTS: We developed an innovative strategy using CRISPR/Cas9 to generate UMSC with B2M deletion (B2M-UMSC). AlloUMSC injection induced CD8+ T cell-mediated immune rejection in immune competent rats, whereas no CD8+ T cell-mediated killing against B2M-UMSC was observed even when the cells were treated with IFN-γ. Moreover, we demonstrate that UMSC-derived exosomes can inhibit cardiac fibrosis and restore cardiac function, and exosomes derived from B2M-UMSC are more efficient than those derived from UMSC, indicating that the beneficial effect of exosomes can be enhanced by modulating exosome's imprinting. Mechanistically, microRNA sequencing identifies miR-24 as a major component of the exosomes from B2M-UMSCs. Bioinformatics analysis identifies Bim as a putative target of miR-24. Loss-of-function studies at the cellular level and gain-of-function approaches in exosomes show that the beneficial effects of B2M-UMSCs are mediated by the exosome/miR-24/Bim pathway. CONCLUSION: Our findings demonstrate that modulation of exosome's imprinting via B2M knockout is an efficient strategy to prevent the immune rejection of alloUMSCs. This study paved the way to the development of new strategies for tissue repair and regeneration without the need for HLA matching.


Subject(s)
CRISPR-Cas Systems/genetics , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Myocardial Infarction/therapy , beta 2-Microglobulin/genetics , Animals , Bcl-2-Like Protein 11/metabolism , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Exosomes/metabolism , Fibrosis/prevention & control , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Rats , beta 2-Microglobulin/metabolism
3.
Carcinogenesis ; 41(6): 841-849, 2020 07 10.
Article in English | MEDLINE | ID: mdl-31560739

ABSTRACT

Chloride intracellular channel protein 4 (CLIC4) has been implicated in different types of cancers, but the role of CLIC4 in the development of gastric cancer (GC) remains unknown. We analyzed the expression of CLIC4 in 102 pairs of gastric adenocarcinomas by western blot and real-time PCR. Our data revealed that the expression of CLIC4 is reduced in GC tumor tissues compared with adjacent normal tissues. The expression levels of CLIC4 correlate inversely with the clinical stage of GC. CLIC4 expression is lowest in MKN45 cells, which have the highest tumorigenic potential and express the highest levels of cancer stem cell markers CD44 and OCT4, compared with N87 and AGS cells. Exogenous overexpression of CLIC4 downregulated the expression of CD44 and OCT4, and inhibited migration, invasion and epithelial-mesenchymal transition (EMT). Moreover, anchorage-independent growth of GC cells was decreased and the cells became more sensitive to 5-fluorouracil and etoposide treatment when CLIC4 was overexpressed. The ability of N87 cells to form tumors in nude mice was enhanced when CLIC4 was silenced. We, for the first time, demonstrate that CLIC4 suppresses tumor growth by inhibiting cancer cell stemness and EMT.


Subject(s)
Biomarkers, Tumor/metabolism , Chloride Channels/antagonists & inhibitors , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Chloride Channels/genetics , Chloride Channels/metabolism , Female , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prognosis , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Cell Mol Med ; 24(1): 695-710, 2020 01.
Article in English | MEDLINE | ID: mdl-31729180

ABSTRACT

Generating universal human umbilical mesenchymal stem cells (UMSCs) without immune rejection is desirable for clinical application. Here we developed an innovative strategy using CRISPR/Cas9 to generate B2M- UMSCs in which human leucocyte antigen (HLA) light chain ß2-microglobulin (B2M) was deleted. The therapeutic potential of B2M- UMSCs was examined in a mouse ischaemic hindlimb model. We show that B2M- UMSCs facilitated perfusion recovery and enhanced running capability, without inducing immune rejection. The beneficial effect was mediated by exosomes. Mechanistically, microRNA (miR) sequencing identified miR-24 as a major component of the exosomes originating from B2M- UMSCs. We identified Bim as a potential target of miR-24 through bioinformatics analysis, which was further confirmed by loss-of-function and gain-of-function approaches. Taken together, our data revealed that knockout of B2M is a convenient and efficient strategy to prevent UMSCs-induced immune rejection, and it provides a universal clinical-scale cell source for tissue repair and regeneration without the need for HLA matching in the future.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Exosomes/metabolism , Hindlimb/cytology , Ischemia/prevention & control , MicroRNAs/genetics , Stem Cell Transplantation/adverse effects , beta 2-Microglobulin/physiology , Animals , Bcl-2-Like Protein 11/genetics , Exosomes/genetics , Hindlimb/immunology , Hindlimb/injuries , Hindlimb/metabolism , Humans , Ischemia/etiology , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/administration & dosage , Stem Cells/metabolism , Stem Cells/pathology , Umbilical Cord/metabolism , Umbilical Cord/pathology
5.
Br J Cancer ; 122(10): 1477-1485, 2020 05.
Article in English | MEDLINE | ID: mdl-32203224

ABSTRACT

BACKGROUND: DAXX is a transcription repressor that has been implicated in several types of cancers, but its role in the development of gastric cancer remains unknown. METHODS: We analysed the expression of DAXX in 83 pairs of gastric cancer samples, including neoplastic and adjacent tissues, and correlated the expression levels with clinical stages. We also investigated the molecular mechanisms by which DAXX downregulation promotes cancer growth using both in vitro and in vivo models. RESULTS: DAXX was downregulated in advanced gastric cancer samples. The expression of DAXX inversely correlates with that of cancer stem cell markers CD44 and Oct4 in gastric cancer lines. DAXX overexpression in gastric cancer cells inhibited migration, invasion and epithelial- mesenchymal transition (EMT). The inhibition of EMT was achieved through the repression of SNAI3, a key inducer of EMT, by recruiting HDAC-1 into the nucleus. Using a xenograft mouse model, we demonstrated that the MKN45 cells formed smaller tumours when DAXX was overexpressed. Wild-type AGS cells were not able to form tumours in nude mice, but in contrast, formed visible tumours when DAXX was silenced in the cells. CONCLUSION: We for the first time demonstrated that DAXX functions as a tumour suppressor in gastric cancer by inhibiting stem cell growth and EMT.


Subject(s)
Co-Repressor Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Molecular Chaperones/genetics , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/genetics , Adult , Aged , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Male , Mice , Middle Aged , Neoplastic Stem Cells/pathology , Stomach Neoplasms/pathology
6.
Chem Biodivers ; 17(1): e1900534, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31730730

ABSTRACT

Brassica rapa L., also called NIUMA, is used empirically in Tibetan medicine for its antioxidant, anti-inflammatory and antiradiation activities. This study explored the hepatoprotective effects of B. rapa polysaccharides (BRPs) on acute liver injury induced by carbon tetrachloride (CCl4 ) in mice and the underlying mechanisms. Mice were treated with CCl4 after the oral administration of BRPs (55, 110 and 220 mg/kg) or bifendate (100 mg/kg) for 7 days. Blood and liver samples of mice were collected for analysis after 24 h. The ALP, ALT and AST levels and the biological activities of SOD, MDA and GSH-Px were measured. Histopathological changes in the liver were determined through hematoxylin and eosin staining. Moreover, TNF-α, IL-1ß and IL-6 expression levels were detected by commercial reagent kits. Finally, Western blot analysis was used to check the relative expression levels of caspase-3, p-JAK2 and p-STAT3. The BRP pre-treatment significantly decreased the enzymatic activities of ALT, ALP and AST in the serum, markedly increased the activities of SOD and GSH-Px in the liver and reduced the MDA concentration in the liver. BRPs alleviated hepatocyte injury and markedly inhibited the expression of TNF-α, IL-1ß and IL-6, also downregulating the CCl4 -induced hepatic tissue expression of caspase-3. Furthermore, BRPs inhibited the JAK2/STAT3 signaling pathway in a dose-dependent manner in the liver. This study demonstrated that BRPs exert hepatoprotective effect against the CCl4 -induced liver injury via modulating the apoptotic and inflammatory responses and downregulating the JAK2/STAT3 signaling pathway. Therefore, B. rapa could be considered a hepatoprotective medicine.


Subject(s)
Apoptosis/drug effects , Brassica rapa/chemistry , Chemical and Drug Induced Liver Injury/drug therapy , Cytokines/analysis , Inflammation/drug therapy , Oxidative Stress/drug effects , Polysaccharides/pharmacology , Animals , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cytokines/antagonists & inhibitors , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mice, Inbred Strains , Polysaccharides/administration & dosage
7.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29793952

ABSTRACT

Cytosolic DNA arising from intracellular pathogens is sensed by cyclic GMP-AMP synthase (cGAS) and triggers a powerful innate immune response. However, herpes simplex virus 1 (HSV-1), a double-stranded DNA virus, has developed multiple mechanisms to attenuate host antiviral machinery and facilitate viral infection and replication. In the present study, we found that HSV-1 tegument protein VP22 acts as an inhibitor of cGAS/stimulator of interferon genes (cGAS/STING)-mediated production of interferon (IFN) and its downstream antiviral genes. Our results showed that ectopic expression of VP22 decreased cGAS/STING-mediated IFN-ß promoter activation and IFN-ß production. Infection with wild-type (WT) HSV-1, but not VP22-deficient virus (ΔVP22), inhibited immunostimulatory DNA (ISD)-induced activation of the IFN signaling pathway. Further study showed that VP22 interacted with cGAS and inhibited the enzymatic activity of cGAS. In addition, stable knockdown of cGAS facilitated the replication of ΔVP22 virus but not the WT. In summary, our findings indicate that HSV-1 VP22 acts as an antagonist of IFN signaling to persistently evade host innate antiviral responses.IMPORTANCE cGAS is very important for host defense against viral infection, and many viruses have evolved ways to target cGAS and successfully evade the attack by the immune system of their susceptible host. This study demonstrated that HSV-1 tegument protein VP22 counteracts the cGAS/STING-mediated DNA-sensing antiviral innate immunity signaling pathway by inhibiting the enzymatic activity of cGAS. The findings in this study will expand our understanding of the interaction between HSV-1 replication and the host DNA-sensing signaling pathway.


Subject(s)
Herpesvirus 1, Human/immunology , Immunity, Innate , Membrane Proteins/immunology , Nucleotidyltransferases/immunology , Signal Transduction/immunology , Viral Structural Proteins/immunology , Animals , Chlorocebus aethiops , HEK293 Cells , Herpesvirus 1, Human/genetics , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Membrane Proteins/genetics , Nucleotidyltransferases/genetics , Signal Transduction/genetics , Vero Cells , Viral Structural Proteins/genetics
9.
Biochem Biophys Res Commun ; 503(4): 2993-2997, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30115379

ABSTRACT

RUNX1 is a transcription factor that is not expressed in uninjured muscles, but can be detected in denervated muscles, suggesting a role of RUNX1 in muscle's response to injury. However, the role of RUNX1 in muscle's response to ischemia has not been reported. Our study showed that Runx1 is up regulated in skeletal muscle during ischemia reperfusion induced injury. Over-expression of Runx1 in C2C12 cells inhibits myogenic differentiation, but promotes proliferation of myoblasts. Consistent with these findings, we found that Runx1 expression was decreased in differentiated satellite cells. Our results indicate that Runx1 regulates muscle regeneration by promoting proliferation of satellite cells.


Subject(s)
Cell Proliferation , Core Binding Factor Alpha 2 Subunit/physiology , Ischemia , Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Animals , Cell Differentiation , Cell Line , Mice , Muscle Development , Myoblasts
10.
Adv Exp Med Biol ; 1088: 347-368, 2018.
Article in English | MEDLINE | ID: mdl-30390260

ABSTRACT

Muscle atrophy in aging is characterized by progressive loss of muscle mass and function. Muscle mass is determined by the balance of synthesis and degradation of protein, which are regulated by several signaling pathways such as ubiquitin-proteasome system, autophagy-lysosome systems, oxidative stress, proinflammatory cytokines, hormones, and so on. Sufficient nutrition can enhance protein synthesis, while exercise can improve the quality of life in the elderly. This chapter will discuss the epidemiology, pathogenesis, as well as the current treatment for aging-induced muscular atrophy.


Subject(s)
Aging/pathology , Muscle, Skeletal/pathology , Muscular Atrophy/physiopathology , Autophagy , Cytokines/physiology , Humans , Muscle Proteins , Muscular Atrophy/epidemiology , Muscular Atrophy/therapy , Oxidative Stress , Proteasome Endopeptidase Complex/physiology , Signal Transduction , Ubiquitin/physiology
11.
Appl Microbiol Biotechnol ; 101(4): 1739-1751, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27858136

ABSTRACT

Mine tailings contain high concentrations of metal contaminants and only little nutrients, making the tailings barren for decades after the mining has been terminated. Effective phytoremediation of mine tailings calls for deep-rooted, metal accumulating, and soil fertility increasing plants with tolerance against harsh environmental conditions. We assessed the potential of the biofuel leguminous tree Pongamia pinnata inoculated with plant growth promoting rhizobia to remediate iron-vanadium-titanium oxide (V-Ti magnetite) mine tailing soil by pot experiment and in situ remediation test. A metal tolerant rhizobia strain PZHK1 was isolated from the tailing soil and identified as Bradyrhizobium liaoningense by phylogenetic analysis. Inoculation with PZHK1 increased the growth of P. pinnata both in V-Ti magnetite mine tailings and in Ni-contaminated soil. Furthermore, inoculation increased the metal accumulation capacity and superoxide dismutase activity of P. pinnata. The concentrations of Ni accumulated by inoculated plants were higher than the hyperaccumulator threshold. Inoculated P. pinnata accumulated high concentration of Fe, far exceeding the upper limit (1000 mg kg-1) of Fe in plant tissue. In summary, P. pinnata-B. liaoningense PZHK1 symbiosis showed potential to be applied as an effective phytoremediation technology for mine tailings and to produce biofuel feedstock on the marginal land.


Subject(s)
Bradyrhizobium/metabolism , Mining , Biodegradation, Environmental
12.
Adv Exp Med Biol ; 1000: 333-353, 2017.
Article in English | MEDLINE | ID: mdl-29098629

ABSTRACT

It is known that moderate exercise can prevent the development of cardiovascular diseases, but the exact molecular mechanisms mediating cardioprotective effect of exercise remain unknown. Emerging evidence suggests that exercise has great impact on the biogenesis of exosomes, which have been found in both interstitial fluid and circulation, and play important roles in cellular communication. Exosomes carry functional molecules such as mRNAs, microRNA, and specific proteins, which can be used in the early diagnosis and targeted therapy of a variety of diseases. Our review focus on the current knowledge on exosome production, secretion, uptake and how exercise influence exosome content. We also highlight recent research development in exosome based approach for cardiac repair.


Subject(s)
Cardiovascular Diseases/physiopathology , Exercise/physiology , Exosomes/genetics , MicroRNAs/genetics , Adaptation, Physiological/genetics , Adaptation, Physiological/physiology , Animals , Biomarkers/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Exosomes/metabolism , Gene Expression Regulation , Humans
13.
Adv Exp Med Biol ; 998: 187-206, 2017.
Article in English | MEDLINE | ID: mdl-28936741

ABSTRACT

Cardiovascular diseases resulting from ischemic heart diseases remain to be the main causes of heart failure and death despite significant advances in medical treatment. The development of new therapies for heart failure is thus required to improve the outcome in these patients, and this has led to the development of cell-based therapies. Animal studies showed interesting results using various cell types. Some stem cell based therapies have been tested in clinical trials. Although the results were encouraging, challenges remain. Tumorigenic potential, immune rejection, and low engraftment and survival rate of transplant cells have hindered the widespread application of stem cells in the clinic. Fortunately, exosome based therapy could avoid these problems associated with cell therapy. Future research should focus on how various molecules are sorted into exosomes and this information will help to design better exosomes for treatment of cardiovascular diseases. Recent studies suggest that exosome content can vary depending on how cells are challenged. It would be important to find out exactly what types of cellular stress is needed for producing most useful exosomes. Alternatively, specific molecules can be introduced into exosomes by genetic engineering in order to treat specific conditions and to improve efficacy.


Subject(s)
Cardiovascular Diseases/surgery , Embryonic Stem Cells/transplantation , Exosomes/transplantation , Myocardium/pathology , Myocytes, Cardiac/transplantation , Regeneration , Stem Cell Transplantation , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Embryonic Stem Cells/metabolism , Exosomes/genetics , Exosomes/metabolism , Exosomes/pathology , Gene Expression Regulation , Humans , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recovery of Function , Signal Transduction , Stem Cell Transplantation/adverse effects
16.
J Cell Physiol ; 229(5): 580-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24122208

ABSTRACT

Despite the fact that tissue engineered heart valves (TEHV) hold great promise for heart valve disease treatment, one of the challenges is to find suitable seeding cells. Bone marrow derived mesenchymal stem cells (MSCs) were considered to be one of the best seed cell sources. In this study we propose a novel approach to promote stem cell differentiation into the seed cells of TEHV, valvular interstitial cells (VICs). Newly induced MSCs (iMSCs) were created from a co-culture niche in which healthy human donor derived MSCs were co-cultured with cardiac fibroblasts (H9C2 cell line). Then iMSCs were transfected with either a mock vector (iMSCs(mock) ) as controls or with a vector that overexpresses thefibroblast inducible factor 14 (Fn14) gene (iMSCs(Fn14) ). Immunofluorescence staining was performed to assay VIC differentiation. Western blot analysis was performed to analyze the involved signaling pathway. The results demonstrate that the expression of α-smooth muscle actin (SMA) was significantly higher in iMSCs(Fn14) as compared with iMSC(mock) , and MSC, and also had higher co-alignment of α-actinin and stress fiber (F-actin) in bundles. Additionally, increased biosynthesis of extracellular matrix (ECM) proteins including collagen I, collagen III, and fibronection were observed in iMSCs(Fn14) in comparison with iMSCs(mock) . These data observed in iMSCs(Fn14) were in accordance with VIC phenotype from normal heart valves. In addition, the PI3K/Akt signaling pathway was activated in iMSCs(Fn14) which allowed higher Akt phosphorylation (p-Akt) levels and SMA levels, whereas, it was attenuated by LY294002 (PI3K/Akt inhibitor). These new findings of the effect of Fn14 on VIC-like cell differentiation may provide a novel therapeutic strategy for heart valve disease treatment.


Subject(s)
Cell Differentiation/physiology , Heart Valves/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Cell Line , Coculture Techniques , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation/physiology , Humans , Mesenchymal Stem Cells/physiology , Phosphoinositide-3 Kinase Inhibitors , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/physiology , TWEAK Receptor , Tissue Engineering , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
Theranostics ; 14(10): 3927-3944, 2024.
Article in English | MEDLINE | ID: mdl-38994017

ABSTRACT

Rationale: Myocardial infarction (MI) is a severe global clinical condition with widespread prevalence. The adult mammalian heart's limited capacity to generate new cardiomyocytes (CMs) in response to injury remains a primary obstacle in developing effective therapies. Current approaches focus on inducing the proliferation of existing CMs through cell-cycle reentry. However, this method primarily elevates cyclin dependent kinase 6 (CDK6) and DNA content, lacking proper cytokinesis and resulting in the formation of dysfunctional binucleated CMs. Cytokinesis is dependent on ribosome biogenesis (Ribo-bio), a crucial process modulated by nucleolin (Ncl). Our objective was to identify a novel approach that promotes both DNA synthesis and cytokinesis. Methods: Various techniques, including RNA/protein-sequencing analysis, Ribo-Halo, Ribo-disome, flow cytometry, and cardiac-specific tumor-suppressor retinoblastoma-1 (Rb1) knockout mice, were employed to assess the series signaling of proliferation/cell-cycle reentry and Ribo-bio/cytokinesis. Echocardiography, confocal imaging, and histology were utilized to evaluate cardiac function. Results: Analysis revealed significantly elevated levels of Rb1, bur decreased levels of circASXL1 in the hearts of MI mice compared to control mice. Deletion of Rb1 induces solely cell-cycle reentry, while augmenting the Ribo-bio modulator Ncl leads to cytokinesis. Mechanically, bioinformatics and the loss/gain studies uncovered that circASXL1/CDK6/Rb1 regulates cell-cycle reentry. Moreover, Ribo-Halo, Ribo-disome and circRNA pull-down assays demonstrated that circASXL1 promotes cytokinesis through Ncl/Ribo-bio. Importantly, exosomes derived from umbilical cord mesenchymal stem cells (UMSC-Exo) had the ability to enhance cardiac function by facilitating the coordinated signaling of cell-cycle reentry and Ribo-bio/cytokinesis. These effects were attenuated by silencing circASXL1 in UMSC-Exo. Conclusion: The series signaling of circASXL1/CDK6/Rb1/cell-cycle reentry and circASXL1/Ncl/Ribo-bio/cytokinesis plays a crucial role in cardiac repair. UMSC-Exo effectively repairs infarcted myocardium by stimulating CM cell-cycle reentry and cytokinesis in a circASXL1-dependent manner. This study provides innovative therapeutic strategies targeting the circASXL1 signaling network for MI and offering potential avenues for enhanced cardiac repair.


Subject(s)
Cell Cycle , Cytokinesis , Mice, Knockout , Myocardial Infarction , Myocytes, Cardiac , Ribosomes , Animals , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Ribosomes/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Nucleolin , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/genetics , Cell Proliferation , Male , Humans
19.
Signal Transduct Target Ther ; 8(1): 15, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36617563

ABSTRACT

The ribosome is a multi-unit complex that translates mRNA into protein. Ribosome biogenesis is the process that generates ribosomes and plays an essential role in cell proliferation, differentiation, apoptosis, development, and transformation. The mTORC1, Myc, and noncoding RNA signaling pathways are the primary mediators that work jointly with RNA polymerases and ribosome proteins to control ribosome biogenesis and protein synthesis. Activation of mTORC1 is required for normal fetal growth and development and tissue regeneration after birth. Myc is implicated in cancer development by enhancing RNA Pol II activity, leading to uncontrolled cancer cell growth. The deregulation of noncoding RNAs such as microRNAs, long noncoding RNAs, and circular RNAs is involved in developing blood, neurodegenerative diseases, and atherosclerosis. We review the similarities and differences between eukaryotic and bacterial ribosomes and the molecular mechanism of ribosome-targeting antibiotics and bacterial resistance. We also review the most recent findings of ribosome dysfunction in COVID-19 and other conditions and discuss the consequences of ribosome frameshifting, ribosome-stalling, and ribosome-collision. We summarize the role of ribosome biogenesis in the development of various diseases. Furthermore, we review the current clinical trials, prospective vaccines for COVID-19, and therapies targeting ribosome biogenesis in cancer, cardiovascular disease, aging, and neurodegenerative disease.


Subject(s)
COVID-19 , Neoplasms , Neurodegenerative Diseases , Humans , Pregnancy , Female , COVID-19 Vaccines/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , COVID-19/metabolism , Ribosomes/genetics , Ribosomal Proteins/genetics , Neoplasms/drug therapy , Neoplasms/genetics , RNA, Untranslated , Mechanistic Target of Rapamycin Complex 1/metabolism
20.
MedComm (2020) ; 4(1): e202, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36636367

ABSTRACT

The regenerative capacity of skeletal muscle is dependent on satellite cells. The circadian clock regulates the maintenance and function of satellite cells. Cryptochrome 2 (CRY2) is a critical component of the circadian clock, and its role in skeletal muscle regeneration remains controversial. Using the skeletal muscle lineage and satellite cell-specific CRY2 knockout mice (CRY2scko), we show that the deletion of CRY2 enhances muscle regeneration. Single myofiber analysis revealed that deletion of CRY2 stimulates the proliferation of myoblasts. The differentiation potential of myoblasts was enhanced by the loss of CRY2 evidenced by increased expression of myosin heavy chain (MyHC) and myotube formation in CRY2-/- cells versus CRY2+/+ cells. Immunostaining revealed that the number of mononucleated paired box protein 7 (PAX7+) cells associated with myotubes formed by CRY2-/- cells was increased compared with CRY2+/+ cells, suggesting that more reserve cells were produced in the absence of CRY2. Loss of CRY2 leads to the activation of the ERK1/2 signaling pathway and ETS1, which binds to the promoter of PAX7 to induce its transcription. CRY2 deficient myoblasts survived better in ischemic muscle. Therefore, CRY2 is essential in regulating skeletal muscle repair.

SELECTION OF CITATIONS
SEARCH DETAIL