Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Theranostics ; 14(6): 2544-2559, 2024.
Article in English | MEDLINE | ID: mdl-38646641

ABSTRACT

Background: Mechanical forces are indispensable for bone healing, disruption of which is recognized as a contributing cause to nonunion or delayed union. However, the underlying mechanism of mechanical regulation of fracture healing is elusive. Methods: We used the lineage-tracing mouse model, conditional knockout depletion mouse model, hindlimb unloading model and single-cell RNA sequencing to analyze the crucial roles of mechanosensitive protein polycystin-1 (PC1, Pkd1) promotes periosteal stem/progenitor cells (PSPCs) osteochondral differentiation in fracture healing. Results: Our results showed that cathepsin (Ctsk)-positive PSPCs are fracture-responsive and mechanosensitive and can differentiate into osteoblasts and chondrocytes during fracture repair. We found that polycystin-1 declines markedly in PSPCs with mechanical unloading while increasing in response to mechanical stimulus. Mice with conditional depletion of Pkd1 in Ctsk+ PSPCs show impaired osteochondrogenesis, reduced cortical bone formation, delayed fracture healing, and diminished responsiveness to mechanical unloading. Mechanistically, PC1 facilitates nuclear translocation of transcriptional coactivator TAZ via PC1 C-terminal tail cleavage, enhancing osteochondral differentiation potential of PSPCs. Pharmacological intervention of the PC1-TAZ axis and promotion of TAZ nuclear translocation using Zinc01442821 enhances fracture healing and alleviates delayed union or nonunion induced by mechanical unloading. Conclusion: Our study reveals that Ctsk+ PSPCs within the callus can sense mechanical forces through the PC1-TAZ axis, targeting which represents great therapeutic potential for delayed fracture union or nonunion.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Chondrocytes , Fracture Healing , Osteogenesis , Stem Cells , TRPP Cation Channels , Animals , Fracture Healing/physiology , Mice , TRPP Cation Channels/metabolism , TRPP Cation Channels/genetics , Chondrocytes/metabolism , Stem Cells/metabolism , Osteogenesis/physiology , Mice, Knockout , Chondrogenesis/physiology , Periosteum/metabolism , Osteoblasts/metabolism , Osteoblasts/physiology , Disease Models, Animal , Male
2.
Front Endocrinol (Lausanne) ; 13: 899731, 2022.
Article in English | MEDLINE | ID: mdl-36060945

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), hallmarked by liver steatosis, is becoming a global concern, but effective and safe drugs for NAFLD are still lacking at present. Parathyroid hormone (PTH), the only FDA-approved anabolic treatment for osteoporosis, is important in calcium-phosphate homeostasis. However, little is known about its potential therapeutic effects on other diseases. Here, we report that intermittent administration of PTH ameliorated non-alcoholic liver steatosis in diet-induced obese (DIO) mice and db/db mice, as well as fasting-induced hepatic steatosis. In vitro, PTH inhibits palmitic acid-induced intracellular lipid accumulation in a parathyroid hormone 1 receptor (PTH1R)-dependent manner. Mechanistically, PTH upregulates the expression of genes involved in lipid ß-oxidation and suppresses the expression of genes related to lipid uptake and de novo lipogenesis by activating the cAMP/PKA/CREB pathway. Taken together, our current finding proposes a new therapeutic role of PTH on NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Parathyroid Hormone , Animals , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Lipids , Lipogenesis , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Parathyroid Hormone/metabolism , Parathyroid Hormone/therapeutic use , Receptor, Parathyroid Hormone, Type 1/metabolism
3.
Cell Prolif ; 54(8): e13095, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34254370

ABSTRACT

OBJECTIVES: Scavenger receptor class A, member 3 (Scara3) was involved in adipogenesis. However, the effect of Scara3 on the switch between osteogenesis and adipogenesis of bone marrow mesenchymal stem cells (BMSCs) remains elusive. MATERIALS AND METHODS: The correlations between SCARA3 with the osteogenic-related were analysed based on the GTEx database. The effects of Scara3 on osteogenic or adipogenic differentiation of BMSCs were evaluated by qPCR, Western blot (WB) and cell staining. The mechanisms of Scara3 regulating Foxo1 and autophagy were validated by co-expression analysis, WB and immunofluorescence. In vivo, Scara3 adeno-associated virus was injected into intra-bone marrow of the aged mice and ovariectomized (OVX) mice whose phenotypes were confirmed by micro-CT, calcein double labelling and immunochemistry (HE and OCN staining). RESULTS: SCARA3 was positively correlated with osteogenic-related genes. Scara3 expression gradually decreased during adipogenesis but increased during osteogenesis. Moreover, the deletion of Scara3 favoured adipogenesis over osteogenesis, whereas overexpression of Scara3 significantly enhanced the osteogenesis at the expense of adipogenesis. Mechanistically, Scara3 controlled the cell fate by promoting Foxo1 expression and autophagy flux. In vivo, Scara3 promoted bone formation and reduced bone marrow fat accumulation in OVX mice. In the aged mice, Scara3 overexpression alleviated bone loss as well. CONCLUSIONS: This study suggested that Scara3 regulated the switch between adipocyte and osteoblast differentiation, which represented a potential therapeutic target for bone loss and osteoporosis.


Subject(s)
Adipocytes/cytology , Forkhead Box Protein O1/metabolism , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Scavenger Receptors, Class A/metabolism , Adipocytes/metabolism , Adipogenesis , Aging , Animals , Autophagy , Cell Differentiation , Cells, Cultured , Female , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis , RNA Interference , RNA, Small Interfering/metabolism , Scavenger Receptors, Class A/antagonists & inhibitors , Scavenger Receptors, Class A/genetics
4.
Cell Prolif ; 52(4): e12624, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31038249

ABSTRACT

OBJECTIVES: Excessive oxidative stress and diminished antioxidant defences could contribute to age-related tissue damage and various diseases including age-related osteoporosis. Dendrobium officinale polysaccharides (DOPs), a major ingredient from a traditional Chinese medicine, have a great potential of antioxidative activity. In this study, we explore the role of DOP in age-related osteoporosis that remains elusive. MATERIALS AND METHODS: Oxidative stimulation and DOP were used to treat bone marrow mesenchymal stem cells (BMSCs), whose lineage commitment was measured by adipogenic- and osteoblastic-induced differentiation analysis. The oxidative stress and antioxidant capacity of BMSCs under the treatment of DOP were analysed by the level of MDA, SOD. Related mechanism studies were confirmed by qRT-PCR, Western blotting and siRNA transfection. DOP was orally administrated in aged mice whose phenotype was confirmed by micro-CT, immunofluorescence, immunochemistry and calcein double-labelling analysis. RESULTS: Dendrobium officinale polysaccharide treatment markedly increased osteogenic differentiation of BMSCs, while inhibiting adipogenic differentiation. In vitro, DOP could rescue H2O2-induced switch of BMSCs differentiation fate. However, this effect was abolished in BMSCs when interfered with Nrf2 siRNA. Furthermore, administration of DOP to aged mice significantly increased the bone mass and reduced the marrow adipose tissue (MAT) accompanied with decreased oxidative stress of BMSCs. CONCLUSIONS: Our study reveals that DOP can attenuate bone loss and MAT accumulation through NRF2 antioxidant signalling, which may represent as potential therapeutic agent for age-related osteoporosis.


Subject(s)
Adipogenesis/drug effects , Cell Differentiation/drug effects , Dendrobium/chemistry , Osteogenesis/drug effects , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Adult , Aged , Animals , Antioxidants/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Middle Aged , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Young Adult
5.
Biomed Res Int ; 2015: 624037, 2015.
Article in English | MEDLINE | ID: mdl-26221600

ABSTRACT

We previously reported that Runx2/miR-3960/miR-2861 regulatory feedback loop stimulates osteoblast differentiation. However, the effect of this feedback loop on the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) remains unclear. Our recent study showed that miR-2861 and miR-3960 expression increases significantly during ß-glycerophosphate-induced osteogenic transdifferentiation of VSMCs. Overexpression of miR-2861 or miR-3960 in VSMCs enhances ß-glycerophosphate-induced osteoblastogenesis, whereas inhibition of miR-2861 or miR-3960 expression attenuates it. MiR-2861 or miR-3960 promotes osteogenic transdifferentiation of VSMCs by targeting histone deacetylase 5 or Homeobox A2, respectively, resulting in increased runt-related transcription factor 2 (Runx2) protein production. Furthermore, overexpression of Runx2 induces miR-2861 and miR-3960 transcription, and knockdown of Runx2 attenuates ß-glycerophosphate-induced miR-2861 and miR-3960 transcription in VSMCs. Thus, our data show that Runx2/miR-3960/miR-2861 positive feedback loop plays an important role in osteogenic transdifferentiation of VSMCs and contributes to vascular calcification.


Subject(s)
Cell Transdifferentiation/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Feedback, Physiological , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Osteogenesis/genetics , Animals , Cell Transdifferentiation/drug effects , Feedback, Physiological/drug effects , Gene Expression Regulation/drug effects , Glycerophosphates/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Transcription, Genetic/drug effects
6.
J Clin Invest ; 125(4): 1509-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751060

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) exhibit an age-dependent reduction in osteogenesis that is accompanied by an increased propensity toward adipocyte differentiation. This switch increases adipocyte numbers and decreases the number of osteoblasts, contributing to age-related bone loss. Here, we found that the level of microRNA-188 (miR-188) is markedly higher in BMSCs from aged compared with young mice and humans. Compared with control mice, animals lacking miR-188 showed a substantial reduction of age-associated bone loss and fat accumulation in bone marrow. Conversely, mice with transgenic overexpression of miR-188 in osterix+ osteoprogenitors had greater age-associated bone loss and fat accumulation in bone marrow relative to WT mice. Moreover, using an aptamer delivery system, we found that BMSC-specific overexpression of miR-188 in mice reduced bone formation and increased bone marrow fat accumulation. We identified histone deacetylase 9 (HDAC9) and RPTOR-independent companion of MTOR complex 2 (RICTOR) as the direct targets of miR-188. Notably, BMSC-specific inhibition of miR-188 by intra-bone marrow injection of aptamer-antagomiR-188 increased bone formation and decreased bone marrow fat accumulation in aged mice. Together, our results indicate that miR-188 is a key regulator of the age-related switch between osteogenesis and adipogenesis of BMSCs and may represent a potential therapeutic target for age-related bone loss.


Subject(s)
Adipocytes/cytology , Aging/genetics , Bone Marrow Cells/cytology , MicroRNAs/physiology , Osteoblasts/cytology , Osteogenesis/physiology , Osteoporosis/prevention & control , 3' Untranslated Regions/genetics , Adipose Tissue/cytology , Aging/metabolism , Animals , Aptamers, Nucleotide/pharmacology , Base Sequence , Bone Density/genetics , Bone Density/physiology , Bone Marrow Cells/metabolism , Bone Remodeling/physiology , Carrier Proteins/antagonists & inhibitors , Cell Differentiation/genetics , Histone Deacetylases , Humans , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/analysis , MicroRNAs/genetics , Molecular Sequence Data , Osteocalcin/analysis , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/physiopathology , Rapamycin-Insensitive Companion of mTOR Protein , Repressor Proteins/antagonists & inhibitors , Sp7 Transcription Factor , Transcription Factors/physiology
7.
J Biol Chem ; 286(14): 12328-39, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21324897

ABSTRACT

Our recent study showed that miR-2861 promotes osteoblast differentiation by targeting histone deacetylase 5, resulting in increased runt-related transcription factor 2 (Runx2) protein production. Here we identified another new microRNA (miRNA) (miR-3960) that played a regulatory role in osteoblast differentiation through a regulatory feedback loop with miR-2861. miR-3960 and miR-2861 were found clustered at the same loci. miR-3960 was transcribed during bone morphogenic protein 2 (BMP2)-induced osteogenesis of ST2 stromal cells. Overexpression of miR-3960 promoted BMP2-induced osteoblastogenesis. However, the inhibition of miR-3960 expression attenuated the osteoblastogenesis. Homeobox A2 (Hoxa2), a repressor of Runx2 expression, was confirmed to be a target of miR-3960. Electrophoretic mobility shift assay and chromatin immunoprecipitation experiments confirmed that Runx2 bound to the promoter of the miR-3960/miR-2861 cluster. Furthermore, overexpression of Runx2 induced miR-3960/miR-2861 transcription, and block of Runx2 expression attenuated BMP2-induced miR-3960/miR-2861 transcription. Here we report that miR-3960 and miR-2861, transcribed together from the same miRNA polycistron, both function in osteoblast differentiation through a novel Runx2/miR-3960/miR-2861 regulatory feedback loop. Our findings provide new insights into the roles of miRNAs in osteoblast differentiation.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/physiology , Osteoblasts/cytology , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Chromatin Immunoprecipitation , Computational Biology , Core Binding Factor Alpha 1 Subunit/genetics , Electrophoretic Mobility Shift Assay , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Osteoblasts/metabolism , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...