Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Front Bioeng Biotechnol ; 12: 1356135, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38600948

RESUMO

Introduction: Developmental engineering based on endochondral ossification has been proposed as a potential strategy for repairing of critical bone defects. Bone development is driven by growth plate-mediated endochondral ossification. Under physiological conditions, growth plate chondrocytes undergo compressive forces characterized by micro-mechanics, but the regulatory effect of micro-mechanical loading on endochondral bone formation has not been investigated. Methods: In this study, a periodic static compression (PSC) model characterized by micro-strain (with 0.5% strain) was designed to clarify the effects of biochemical/mechanical cues on endochondral bone formation. Hydrogel scaffolds loaded with bone marrow mesenchymal stem cells (BMSCs) were incubated in proliferation medium or chondrogenic medium, and PSC was performed continuously for 14 or 28 days. Subsequently, the scaffold pretreated for 28 days was implanted into rat femoral muscle pouches and femoral condylar defect sites. The chondrogenesis and bone defect repair were evaluated 4 or 10 weeks post-operation. Results: The results showed that PSC stimulation for 14 days significantly increased the number of COL II positive cells in proliferation medium. However, the chondrogenic efficiency of BMSCs was significantly improved in chondrogenic medium, with or without PSC application. The induced chondrocytes (ichondrocytes) spontaneously underwent hypertrophy and maturation, but long-term mechanical stimulation (loading for 28 days) significantly inhibited hypertrophy and mineralization in ichondrocytes. In the heterotopic ossification model, no chondrocytes were found and no significant difference in terms of mineral deposition in each group; However, 4 weeks after implantation into the femoral defect site, all scaffolds that were subjected to biochemical/mechanical cues, either solely or synergistically, showed typical chondrocytes and endochondral bone formation. In addition, simultaneous biochemical induction/mechanical loading significantly accelerated the bone regeneration. Discussion: Our findings suggest that microstrain mechanics, biochemical cues, and in vivo microenvironment synergistically regulate the differentiation fate of BMSCs. Meanwhile, this study shows the potential of micro-strain mechanics in the treatment of critical bone defects.

2.
J Orthop Translat ; 44: 88-101, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38282752

RESUMO

Background: Mutations in Slc26a2 cause a spectrum of autosomal-recessive chondrodysplasia with a significant and negligible influence on the quality of life. It has been reported that Slc26a2 deficiency triggers the ATF6 branch of the UPR, which may, in turn, activate the negative regulator of the FGFR3 signaling pathway. However, the correlation between the deletion of Slc26a2 and the augmentation of downstream phosphorylation of FGFR3 has not been investigated in vivo. Methods: First, we constructed Slc26a2 and Fgfr3 double knockout mouse lines and observed gross views of the born mice and histological staining of the tibial growth plates. The second approach was to construct tamoxifen-inducible Cre-ERT2 mouse models to replicate SLC26A2-related non-lethal dysplastic conditions. Pharmacological intervention was performed by administering the FGFR3 inhibitor NVP-BGJ398. The effect of NVP-BGJ398 on chondrocytes was assessed by Alcian blue staining, proliferation, apoptosis, and chondrocyte-specific markers and then verified by western blotting for variations in the downstream markers of FGFR3. The growth process was detected using X-rays, micro-CT examination, histomorphometry staining of growth plates, and immunofluorescence. Results: Genetic ablation of Fgfr3 in embryonic Slc26a2-deficient chondrocytes slightly attenuated chondrodysplasia. Subsequently, in the constructed mild dysplasia model, we found that postnatal intervention with Fgfr3 gene in Slc26a2-deficient chondrocytes partially alleviated chondrodysplasia. In chondrocyte assays, NVP-BGJ398 suppressed the defective phenotype of Slc26a2-deficient chondrocytes and restored the phosphorylation downstream of FGFR3 in a concentration-dependent manner. In addition, in vivo experiments showed significant alleviation of impaired chondrocyte differentiation, and micro-CT analysis showed a clear improvement in trabecular bone microarchitectural parameters. Conclusion: Our results suggested that inhibition of FGFR3 signaling pathway overactivation and NVP-BGJ398 has promising therapeutic implications for the development of SLC26A2-related skeletal diseases in humans. The translational potential of this article: Our data provide genetic and pharmacological evidence that targeting FGFR3 signaling via NVP-BGJ398 could be a route for the treatment of SLC26A2-associated skeletal disorders, which promisingly advances translational applications and therapeutic development.

3.
Mol Med Rep ; 28(3)2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37539751

RESUMO

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that, in Fig. 2A on p. 8311, portraying the results of immunostaining experiments for osterix, the 'GIOP' and 'GIOP+TMP (20)' data panels contained overlapping data, such that these images were derived from apparently the same original source, where they were intended to show the results from differently performed experiments. Moreover, in Fig. 3A on p. 8312 showing the results from ALP staining and Alizarin Red S staining experiments, two pairs of apparently overlapping data panels were identified in the Dex 106 M / TMP 50 µM, 100 µM and 200 µM data panels. After having re­examined their original data, the authors have realized that the data featured in Figs. 2A and 3A were assembled incorrectly in these figures. Revised versions of Fig. 2 and 3, now containing replacement data for the experiments shown in Figs. 2A and 3A, are shown on the next page. Note that these errors did not adversely affect either the results or the overall conclusions reported in this study. All the authors agree with the publication of this corrigendum, and are grateful to the Editor of Molecular Medicine Reports for allowing them the opportunity to publish this. They also wish to apologize to the readership of the Journal for any inconvenience caused. [Molecular Medicine Reports 16: 8307­8314, 2017; DOI: 10.3892/mmr.2017.7610].

4.
Sci Transl Med ; 15(710): eadg3983, 2023 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-37611084

RESUMO

Sulfation is a widespread modification of biomolecules that has been incompletely explored to date. Through cross-phenotype meta-analysis of bone mineral density in up to 426,824 genotyped human participants along with phenotypic characterization of multiple mutant mouse lines, we identified a causative role for sulfate transporter solute carrier family 26 member A2 (SLC26A2) deficiency in osteoporosis. Ablation of SLC26A2 in osteoblasts caused severe bone loss and accumulation of immature bone cells and elicited peculiar pericellular matrix (PCM) production characterized by undersulfation coupled with decreased stiffness. These altered chemophysical properties of the PCM disrupted the formation of focal adhesions in osteoblasts. Bulk RNA sequencing and functional assays revealed that the mechanoreciprocal inhibition of focal adhesion kinase (FAK) and Yes1-associated transcriptional regulator (YAP)/WW domain containing transcription regulator 1 (TAZ) signaling impinged osteoblast maturation upon SLC26A2 deficiency. Moreover, pharmacological abrogation of the Hippo kinases and forced wheel-running ameliorated SLC26A2-deficient osteoporosis by promoting YAP/TAZ activity. Analysis of mouse single-cell RNA sequencing data suggested coordination among sulfate metabolism, focal adhesion, and YAP/TAZ activity during osteoblast-to-osteocyte transition. In addition to the SLC26A2-deficient setting, altered FAK and YAP/TAZ signaling was also observed in bone cells of ovariectomized mice and patients with osteoporosis, and pharmacological enforcing of YAP/TAZ activity ameliorated bone loss in ovariectomized mice. Collectively, these data unveil a role for sulfation in the developmental mechanoreciprocity between matrix and osteoblasts, which could be leveraged to prevent bone loss.


Assuntos
Doenças Ósseas Metabólicas , Osteoporose , Humanos , Animais , Camundongos , Osteoblastos , Osteoporose/genética , Densidade Óssea , Bioensaio , Peptídeos e Proteínas de Sinalização Intracelular
5.
Cell Mol Life Sci ; 80(6): 171, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37261512

RESUMO

Imbalance of bone homeostasis induces bone degenerative diseases such as osteoporosis. Hedgehog (Hh) signaling plays critical roles in regulating the development of limb and joint. However, its unique role in bone homeostasis remained largely unknown. Here, we found that canonical Hh signaling pathway was gradually augmented during osteoclast differentiation. Genetic inactivation of Hh signaling in osteoclasts, using Ctsk-Cre;Smof/f conditional knockout mice, disrupted both osteoclast formation and subsequent osteoclast-osteoblast coupling. Concordantly, either Hh signaling inhibitors or Smo/Gli2 knockdown stunted in vitro osteoclast formation. Mechanistically, Hh signaling positively regulated osteoclast differentiation via transactivation of Traf6 and stabilization of TRAF6 protein. Then, we identified connective tissue growth factor (CTGF) as an Hh-regulatory bone formation-stimulating factor derived from osteoclasts, whose loss played a causative role in osteopenia seen in CKO mice. In line with this, recombinant CTGF exerted mitigating effects against ovariectomy induced bone loss, supporting a potential extension of local rCTGF treatment to osteoporotic diseases. Collectively, our findings firstly demonstrate that Hh signaling, which dictates osteoclast differentiation and osteoclast-osteoblast coupling by regulating TRAF6 and CTGF, is crucial for maintaining bone homeostasis, shedding mechanistic and therapeutic insights into the realm of osteoporosis.


Assuntos
Doenças Ósseas Metabólicas , Reabsorção Óssea , Osteoporose , Feminino , Camundongos , Animais , Osteoclastos/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Osteoblastos/metabolismo , Osteogênese , Transdução de Sinais , Osteoporose/genética , Osteoporose/metabolismo , Homeostase , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Diferenciação Celular , Reabsorção Óssea/metabolismo
6.
Matrix Biol ; 110: 76-90, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35472633

RESUMO

Although recent lineage studies strongly support a chondrocyte-to-osteoblast differentiation continuum, the biological significance and molecular basis remain undetermined. In silico analysis at a single-cell level indicates a transient shutdown of Hedgehog-related transcriptome during simulated cartilage-to-bone transition. Prompted by this, we genetically induce gain- and loss-of function to probe the role of Hedgehog signaling in cartilage-to-bone transition. Ablating Smo in hypertrophic chondrocytes (HCs) does not result in any phenotypic outcome, whereas deleting Ptch1 in HCs leads to disrupted formation of primary spongiosa and actively proliferating HCs-derived osteogenic cells that contribute to bony bulges seen in adult mutant mice. In HCs-derived osteoblasts, constitutive activation of Hedgehog signaling blocks their further differentiation to osteocytes. Moreover, ablation of both Smo and Ptch1 in HCs reverses neither persistent Hedgehog signaling nor bone overgrowths. These results establish a functional contribution of extended chondrocyte lineage to bone homeostasis and diseases, governed by an unanticipated mode of regulation for Hedgehog signaling independently of Smo.


Assuntos
Cartilagem , Proteínas Hedgehog , Animais , Diferenciação Celular , Condrócitos , Proteínas Hedgehog/genética , Camundongos , Osteoblastos , Transdução de Sinais
7.
Bioact Mater ; 12: 185-197, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35310379

RESUMO

The technique bottleneck of repairing large bone defects with tissue engineered bone is the vascularization of tissue engineered grafts. Although some studies have shown that extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (BMSCs) promote bone healing and repair by accelerating angiogenesis, the effector molecules and the mechanism remain unclear, which fail to provide ideas for the future research and development of cell-free interventions. Here, we found that Nidogen1-enriched EV (EV-NID1) derived from BMSCs interferes with the formation and assembly of focal adhesions (FAs) by targeting myosin-10, thereby reducing the adhesion strength of rat arterial endothelial cells (RAECs) to the extracellular matrix (ECM), and enhancing the migration and angiogenesis potential of RAECs. Moreover, by delivery with composite hydrogel, EV-NID1 is demonstrated to promote angiogenesis and bone regeneration in rat femoral defects. This study identifies the intracellular binding target of EV-NID1 and further elucidates a novel approach and mechanism, thereby providing a cell-free construction strategy with precise targets for the development of vascularized tissue engineering products.

8.
Mol Med Rep ; 22(6): 4716-4724, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33173955

RESUMO

Osteoporosis is a common disorder characterized by decreased bone mineral density (BMD) and increased fracture risk. The current techniques detect real­time BMD precisely but do not provide adequate information to predict early bone loss. If bone loss could be diagnosed and predicted early, severe osteoporosis and unexpected fractures could be prevented, allowing for an improved quality of life for individuals. In the present study, an ovariectomized rat model of bone loss was established and the serum levels of 78 potential cytokines were determined using a protein array. The BMD of ovariectomized rats was dynamically measured by micro­CT and the early stage of bone loss was defined at the fourth week after surgery. The expression of several serum protein cytokines was indicated to be altered in the ovariectomized rats during an 8­week time­course of bone loss. Linear regression analysis revealed that the serum levels of C­C motif chemokine ligand 2 (CCL2, also known as monocyte chemoattractant protein 1) and C­X­C motif chemokine ligand 1 (CXCL1) were significantly associated with a reduction in BMD. The significance of these two factors in indicating bone mass reduction was further verified by analyzing serum samples from 24 patients with BMD using ELISA and performing a linear regression analysis. The serum levels of CCL2 and CXCL1 were inversely correlated with the bone mass. Therefore, the cytokines CCL2 and CXCL1 may be potential novel predictors of early bone loss and may be clinically relevant for the early diagnosis and prevention of osteoporosis.


Assuntos
Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Osteoporose/diagnóstico , Absorciometria de Fóton , Adolescente , Adulto , Idoso , Animais , Densidade Óssea/fisiologia , Quimiocina CCL2/sangue , Quimiocina CCL2/fisiologia , Quimiocina CXCL1/sangue , Quimiocina CXCL1/fisiologia , Citocinas , Modelos Animais de Doenças , Feminino , Fraturas Ósseas , Humanos , Masculino , Pessoa de Meia-Idade , Osteoporose/metabolismo , Ovariectomia , Ratos , Ratos Sprague-Dawley
9.
Sci Rep ; 10(1): 17127, 2020 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-33033312

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
J Orthop Translat ; 21: 91-99, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32110508

RESUMO

BACKGROUND/OBJECTIVE: Intervertebral disc degeneration (IDD) remains to be an intractable clinical challenge. Although IDD is characterised by loss of notochordal cells (NCs) and dysfunction of nucleus pulposus (NP) cells, little is known about the origin, heterogeneity, fate and maintenance of NCs and NP cells, which further stunts the therapeutic development. Thus, effective tools to spatially and temporally trace specific cell lineage and clarify cell functions in intervertebral disc (IVD) development and homoeostasis are urgently required. METHODS: In this study, NP specimens were obtained from 20 patients with degenerative disc disease or scoliosis. LepR-Cre mice was crossed with R26R-Tdtomato mice to generate LepR-Cre; R26R-Tdtomato mice, which enabled fate-mapping of NPs from embryo stage to late adult. LMNA G609G/G609G mice was used to determine the effect of premature-aging induced IDD on LepR NPs. X-ray imaging was used to measure lumber disc height of mice. RESULTS: Here, we provide the first evidence that the leptin receptor (LepR) is preferentially expressed in NCs at embryonic stages and notochord-derived cells in the postnatal IVD. By using R26R-Tdtomato fluorescent reporter mice, we systematically analysed the specificity of activity and targeting efficiency of leptin receptor-Cre (LepR-Cre) in IVD tissues from the embryonic stage E15.5 to 6-month-old LepR-Cre; Rosa26-Tdtomato (R26R-Tdtomato) mice. Specifically, LepR-Cre targets a distinct subpopulation of notochord-derived cells closely associated with disc homoeostasis. The percentage of LepR-expressing NP cells markedly decreases in the postnatal mouse IVD and, more importantly, in the human IVD with the progression of IDD. Moreover, both spine instability-induced and premature ageing-induced IDD mouse models display the phenotype of IDD with decreased percentage of LepR-expressing NP cells. These findings uncover a potential role of LepR-expressing notochord-derived cells in disc homoeostasis and open the gate for therapeutically targeting the NP cell subpopulation. CONCLUSION: In conclusion, our data prove LepR-Cre mice useful for mapping the fate of specific subpopulations of IVD cells and uncovering the underlying mechanisms of IDD. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: The translation potential of article is that we first identified LepR as a candidate marker of subpopulation of nucleus pulposus (NP) cells and provided LepR as a potential target for the treatment of intervertebral disc degeneration (IDD), which have certain profound significance.

11.
FASEB J ; 34(1): 720-734, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914611

RESUMO

Confusion persists over pathogenesis of spondylolysis. To confirm pathogenicity of the previously identified causative mutation of spondylolysis and investigate the genetic etiology, we generate a new mouse line harboring D673V mutation in the Slc26a2 gene. D673V mutation induces delayed endochondral ossification characterized by transiently reduced chondrocyte proliferation in mice at the early postnatal stage. Adult D673V homozygotes exhibit dysplastic isthmus and reduced bone volume of the dorsal vertebra resembling the detached vertebral bony structure when spondylolysis occurs, including the postzygopophysis, vertebral arch, and spinous process, which causes biomechanical alterations around the isthmic region of L4-5 vertebrae indicated by finite element analysis. Consistently, partial ablation of Slc26a2 in vertebral skeletal cells using Col1a1-Cre; Slc26a2 fl/fl mouse line recapitulates a similar but worsened vertebral phenotype featured by lamellar isthmus. In addition, when reaching late adulthood, D673V homozygotes develop an evident bone-loss phenotype and show impaired osteogenesis. These findings support a multifactorial etiology, involving congenitally predisposed isthmic conditions, altered biomechanics, and age-dependent bone loss, which leads to SLC26A2-related spondylolysis.


Assuntos
Vértebras Lombares/cirurgia , Espondilólise/patologia , Transportadores de Sulfato/efeitos dos fármacos , Envelhecimento , Animais , Vértebras Lombares/patologia , Masculino , Camundongos , Osteogênese/efeitos dos fármacos , Fenótipo , Espondilólise/etiologia , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo
12.
BMC Psychiatry ; 19(1): 281, 2019 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-31510971

RESUMO

BACKGROUND: Prodynorphin (PDYN) gene polymorphisms have been linked with opioid dependence (OD) with conflicting outcomes, the aim of this study is to synthesize the existing evidence of the association between PDYN polymorphisms and OD susceptibility. METHODS: Four databases including PubMed, EMBASE, Web of Science, and Wanfang were retrieved for relevant studies before August, 2018. All identified studies were evaluated using predetermined inclusion and exclusion criteria. Summary odds ratio (OR) and 95% confidence interval (95%CI) were calculated to appraise the association. Statistical analysis was performed using RevMan 5.3 software. RESULTS: A total of seven case-control studies with 3129 cases and 3289 controls were recruited in the meta-analysis. For rs910080, rs1997794, rs1022563, and rs2235749 polymorphisms of PDYN gene, there were six, four, five, and four studies eventually included, respectively. The findings indicated that rs910080 polymorphism was significantly correlated with OD among Asian population under allelic model (A vs. G, OR = 1.30, 95% CI 1.04-1.62, P = 0.02, FDR = 0.05) and dominant model (AA+AG vs. GG, OR = 1.25, 95% CI 1.04-1.51, P = 0.02, FDR = 0.05). However, rs1022563, rs1997794 and rs2235749 polymorphisms did not appear to associate with OD susceptibility. CONCLUSIONS: There existed a significant association between rs1022563 polymorphism and OD among Asian population. As the included studies were not adequate to guarantee a robust and convincing conclusion, future studies with larger sample size among more ethnicities are recommended.


Assuntos
Povo Asiático/genética , Encefalinas/genética , Predisposição Genética para Doença/genética , Transtornos Relacionados ao Uso de Opioides/genética , Polimorfismo de Nucleotídeo Único/genética , Precursores de Proteínas/genética , Estudos de Casos e Controles , Humanos , Transtornos Relacionados ao Uso de Opioides/diagnóstico
13.
Stem Cells Int ; 2019: 5037578, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31089329

RESUMO

Mesenchymal progenitors within bone marrow have multiple differentiation potential and play an essential role in the maintenance of adult skeleton homeostasis. Mesenchymal progenitors located in bone regions other than the bone marrow also display bone-forming properties. However, owing to the differences in each distinct microenvironment, the mesenchymal characteristics of skeletal progenitor cells within different regions of long bones may show some differences. In order to clearly elucidate these differences, we performed a comparative study on mesenchymal progenitors from different regions of long bones. Here, we isolated mesenchymal progenitors from the periosteum, endosteum, and bone marrow of rat long bones. The three groups exhibited similar cellular morphologies and expressed the typical surface markers associated with mesenchymal stem cells. Interestingly, after cell proliferation assays and bidirectional differentiation analysis, periosteal mesenchymal progenitors showed a higher proliferative ability and adipogenic differentiation potential. In contrast, endosteal mesenchymal progenitors were more prone to osteogenic differentiation. Using in vitro osteoclast culture systems, conditioned media from different mesenchymal progenitor cultures were used to induce osteoclastic differentiation. Osteoclast formation was found to be significantly promoted by the secretion of RANKL and IL-6 by endosteal progenitors. Overall, our results provide strong evidence for the importance of selecting the appropriate source of skeletal progenitors for applications in future skeleton regeneration therapies.

14.
EBioMedicine ; 40: 695-709, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30685387

RESUMO

BACKGROUND: Mutations in the SLC26A2 gene cause a spectrum of currently incurable human chondrodysplasias. However, genotype-phenotype relationships of SLC26A2-deficient chondrodysplasias are still perplexing and thus stunt therapeutic development. METHODS: To investigate the causative role of SLC26A2 deficiency in chondrodysplasias and confirm its skeleton-specific pathology, we generated and analyzed slc26a2-/- and Col2a1-Cre; slc26a2fl/fl mice. The therapeutic effect of NVP-BGJ398, an FGFR inhibitor, was tested with both explant cultures and timed pregnant females. FINDINGS: Two lethal forms of human SLC26A2-related chondrodysplasias, achondrogenesis type IB (ACG1B) and atelosteogenesis type II (AO2), are phenocopied by slc26a2-/- mice. Unexpectedly, slc26a2-/- chondrocytes are defective for collagen secretion, exhibiting intracellular retention and compromised extracellular deposition of ColII and ColIX. As a consequence, the ATF6 arm of the unfolded protein response (UPR) is preferentially triggered to overactivate FGFR3 signaling by inducing excessive FGFR3 in slc26a2-/- chondrocytes. Consistently, suppressing FGFR3 signaling by blocking either FGFR3 or phosphorylation of the downstream effector favors the recovery of slc26a2-/- cartilage cultures from impaired growth and unbalanced cell proliferation and apoptosis. Moreover, administration of an FGFR inhibitor to pregnant females shows therapeutic effects on pathological features in slc26a2-/- newborns. Finally, we confirm the skeleton-specific lethality and pathology of global SLC26A2 deletion through analyzing the Col2a1-Cre; slc26a2fl/fl mouse line. INTERPRETATION: Our study unveils a previously unrecognized pathogenic mechanism underlying ACG1B and AO2, and supports suppression of FGFR3 signaling as a promising therapeutic approach for SLC26A2-related chondrodysplasias. FUND: This work was supported by National Natural Science Foundation of China (81871743, 81730065 and 81772377).


Assuntos
Acondroplasia/genética , Acondroplasia/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Transportadores de Sulfato/deficiência , Resposta a Proteínas não Dobradas , Acondroplasia/patologia , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Animais , Cartilagem/metabolismo , Cartilagem/patologia , Diferenciação Celular/genética , Condrócitos/citologia , Condrócitos/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Regulação da Expressão Gênica no Desenvolvimento , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/patologia , Humanos , Camundongos , Camundongos Knockout , Morfogênese/genética , Mutação , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Fenótipo , Resposta a Proteínas não Dobradas/genética
15.
Aging Cell ; 17(3): e12741, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29488314

RESUMO

Aging drives the accumulation of senescent cells (SnCs) including stem/progenitor cells in bone marrow, which contributes to aging-related bone degenerative pathologies. Local elimination of SnCs has been shown as potential treatment for degenerative diseases. As LepR+ mesenchymal stem/progenitor cells (MSPCs) in bone marrow are the major population for forming bone/cartilage and maintaining HSCs niche, whether local elimination of senescent LepR+ MSPCs delays aging-related pathologies and improves local microenvironment need to be well defined. In this study, we performed local delivery of tetramethylpyrazine (TMP) in bone marrow of aging mice, which previously showed to be used for the prevention and treatment of glucocorticoid-induced osteoporosis (GIOP). We found the increased accumulation of senescent LepR+ MSPCs in bone marrow of aging mice, and TMP significantly inhibited the cell senescent phenotype via modulating Ezh2-H3k27me3. Most importantly, local delivery of TMP improved bone marrow microenvironment and maintained bone homeostasis in aging mice by increasing metabolic and anti-inflammatory responses, inducing H-type vessel formation, and maintaining HSCs niche. These findings provide evidence on the mechanisms, characteristics and functions of local elimination of SnCs in bone marrow, as well as the use of TMP as a potential treatment to ameliorate human age-related skeletal diseases and to promote healthy lifespan.


Assuntos
Anti-Inflamatórios/uso terapêutico , Células da Medula Óssea/metabolismo , Células-Tronco Mesenquimais/metabolismo , Pirazinas/uso terapêutico , Vasodilatadores/uso terapêutico , Envelhecimento , Animais , Anti-Inflamatórios/farmacologia , Senescência Celular , Camundongos , Pirazinas/farmacologia , Vasodilatadores/farmacologia
16.
Mol Med Rep ; 16(6): 8307-8314, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28983593

RESUMO

Long­term glucocorticoid therapy results in various side effects, including a high incidence of glucocorticoid­induced osteoporosis (GIOP), which is the most common form of secondary osteoporosis. Excess glucocorticoids reduce the viability of bone marrow­derived mesenchymal stem cells (BMSCs) and prolong osteoclast survival. These two types of cell are essential in the balance between bone formation and resorption. Tetramethylpyrazine (TMP), the pharmacologically active component extracted from Chuanxiong, has been reported to protect BMSCs from glucocorticoid­induced apoptosis. In the present study, the protective effects of TMP on BMSC differentiation and osteoclasts maturation in GIOP were investigated in vivo and in vitro. The immunostaining of osterix (OSX) and tartrate­resistant acid phosphatase (TRAP) staining indicated that TMP promoted osteogenesis and inhibited osteoclastogenesis in a rat model of GIOP. Treatment with 10­6 M dexamethasone (Dex) significantly inhibited BMSC differentiation and increased TRAP­positive cells in vitro. However, different concentrations of TMP (50, 100 and 200 µM) ameliorated the negative effects of Dex by promoting the activity of alkaline phosphatase (ALP) and the calcium mineralization of BMSCs following osteogenic induction, which increased the expression levels of osteogenic genes, including ALP, collagen type I α1, osteocalcin and OSX, and decreased osteoclastogenesis­related genes, including TRAP, nuclear factor of T­cells cytoplasmic 1 and cathepsin K. In addition, it was found that the inhibition of receptor activator of nuclear factor­κB ligand and intereleukin­6 in BMSCs may be a possible mechanism for the protective effects of TMP against glucocorticoid­induced osteoclastogenesis. These results are the first, to the best of our knowledge, to demonstrate that TMP promotes BMSC differentiation and inhibits osteoclastogenesis to ameliorate bone mass change in GIOP.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteogênese/efeitos dos fármacos , Pirazinas/farmacologia , Animais , Reabsorção Óssea , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Feminino , Interleucina-6/genética , Interleucina-6/metabolismo , Osteoclastos/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Ratos , Tomografia Computadorizada por Raios X
17.
Mol Med Rep ; 16(6): 8427-8433, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28983612

RESUMO

Postmenopausal osteoporosis is one of the most prominent worldwide public health problems and the morbidity is increasing with the aging population. It has been demonstrated that early diagnosis and intervention delay the disease progression and improve the outcome. Therefore, searching for biomarkers that are able to identify postmenopausal women at high risk for developing osteoporosis is an effective way to improve the quality of life of patients, and alleviate social and economic burdens. In the present study, a protein array was used to identify potential biomarkers. The bone mineral densities of 10 rats were dynamically measured in an ovariectomized model by micro­computed tomography assessment, and the early stage of osteoporosis was defined. Through the protein array­based screening, the expression levels of six serum protein biomarkers in ovariectomized rats were observed to alter at the initiation stage of the postmenopausal osteoporosis. Fractalkine, tissue inhibitor of metalloproteinases­1 and monocyte chemotactic protein­1 were finally demonstrated to be increased in the serum of eight enrolled postmenopausal osteoporosis patients using ELISA assay and were correlated with the severity of progressive bone loss. These biomarkers may be explored as potential early biomarkers to readily evaluate and diagnose postmenopausal osteoporosis in the clinic.


Assuntos
Proteínas Sanguíneas , Osteoporose Pós-Menopausa/sangue , Osteoporose Pós-Menopausa/diagnóstico , Idoso , Animais , Biomarcadores , Diagnóstico Precoce , Feminino , Humanos , Pessoa de Meia-Idade , Modelos Animais , Ovariectomia/efeitos adversos , Análise Serial de Proteínas , Ratos , Reprodutibilidade dos Testes , Microtomografia por Raio-X
18.
Stem Cells Dev ; 26(6): 419-430, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-27917698

RESUMO

Glucocorticoid-induced osteoporosis (GIOP) is a widespread clinical complication due to the common use of glucocorticoids. Excess glucocorticoids induce apoptosis of bone marrow-derived mesenchymal stem cells (BMSCs), which have been shown to play an increasingly important role in the pathogenesis and therapy of osteoporosis. Tetramethylpyrazine (TMP), an extract from one of the most recognized herbs in traditional Chinese medicine (Chuanxiong), has been reported to have antiapoptotic properties. In this study, we tested whether TMP protects rat BMSCs following exposure to glucocorticoids in vitro and in vivo. We treated BMSCs with different concentrations of TMP (50, 100, or 200 µM) and exposed them to 10-6 M dexamethasone (Dex) for 48 h in vitro. Our data showed that TMP inhibited Dex-induced cytotoxicity and protected BMSCs from apoptosis. Interestingly, further results demonstrated that TMP prevented apoptosis in BMSCs by promoting autophagy in an AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) pathway-dependent manner. In addition, calcein fluorescence double labeling and microcomputed tomography scanning indicated that 12 weeks of TMP administration augmented bone formation and protected trabecular bone mass in GIOP rats. We also discovered that first-passage BMSCs isolated from the TMP treatment group had a lower rate of apoptosis and a higher light chain 3 (LC3)-II/LC3-I ratio than the GIOP group. Our findings demonstrate for the first time that TMP can protect BMSCs from exposure to excess glucocorticoids by promoting autophagy through AMPK/mTOR pathway and might be an effective agent for the prevention and treatment of GIOP.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Osso e Ossos/patologia , Glucocorticoides/efeitos adversos , Células-Tronco Mesenquimais/citologia , Osteoporose/induzido quimicamente , Osteoporose/tratamento farmacológico , Pirazinas/uso terapêutico , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Osso e Ossos/efeitos dos fármacos , Dexametasona , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/patologia , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Pirazinas/farmacologia , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
19.
Sci Rep ; 5: 14080, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26365922

RESUMO

Free fatty acids display diverse effects as signalling molecules through GPCRs in addition to their involvement in cellular metabolism. GPR120, a G protein-coupled receptor for long-chain unsaturated fatty acids, has been reported to mediate adipogenesis in lipid metabolism. However, whether GPR120 also mediates osteogenesis and regulates BMMSCs remain unclear. In this study, we showed that GPR120 targeted the bi-potential differentiation of BMMSCs in a ligand dose-dependent manner. High concentrations of TUG-891 (a highly selective agonist of GPR120) promoted osteogenesis via the Ras-ERK1/2 cascade, while low concentrations elevated P38 and increased adipogenesis. The fine molecular regulation of GPR120 was implemented by up-regulating different integrin subunits (α1, α2 and ß1; α5 and ß3). The administration of high doses of TUG-891 rescued oestrogen-deficient bone loss in vivo, further supporting an essential role of GPR120 in bone metabolism. Our findings, for the first time, showed that GPR120-mediated cellular signalling determines the bi-potential differentiation of BMMSCs in a dose-dependent manner. Additionally, the induction of different integrin subunits was involved in the cytoplasmic regulation of a seesaw-like balance between ERK and p38 phosphorylation. These findings provide new hope for developing novel remedies to treat osteoporosis by adjusting the GPR120-mediated differentiation balance of BMMSCs.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Compostos de Bifenilo/farmacologia , Osso e Ossos/química , Osso e Ossos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Integrinas/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Osteogênese/efeitos dos fármacos , Fenilpropionatos/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo
20.
Nat Commun ; 6: 7112, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26033541

RESUMO

Embryonic stem cells (ESCs) possess pluripotency, which is the capacity of cells to differentiate into all lineages of the mature organism. Increasing evidence suggests that the pluripotent state of ESCs is regulated by a combination of extrinsic and intrinsic factors. The underlying mechanisms, however, are not completely understood. Here, we show that March5, an E3 ubiquitin ligase, is involved in maintaining mouse-ESC (mESC) pluripotency. Knockdown of March5 in mESCs led to differentiation from naive pluripotency. Mechanistically, as a transcriptional target of Klf4, March5 catalyses K63-linked polyubiquitination of Prkar1a, a negative regulatory subunit of PKA, to activate PKA, thereby inhibiting the Raf/MEK/ERK pathway. Moreover, March5 is able to replace a MEK/ERK inhibitor to maintain mESC pluripotency under serum-free culture conditions. In addition, March5 can partially replace the use of Klf4 for somatic cell reprogramming. Collectively, our study uncovers a role for the Klf4-March5-PKA-ERK pathway in maintaining the stemness properties of mESCs.


Assuntos
Diferenciação Celular/genética , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Mitocondriais/genética , Células-Tronco Embrionárias Murinas/metabolismo , Ubiquitina-Proteína Ligases/genética , Animais , Western Blotting , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Imunofluorescência , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Técnicas In Vitro , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Proteínas Mitocondriais/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Quinases raf/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...