Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Genet ; 54(12): 1946-1958, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36456880

RESUMO

Specialized connective tissues, including bone and adipose tissues, control various physiological activities, including mineral and energy homeostasis. However, the identity of stem cells maintaining these tissues throughout adulthood remains elusive. By conducting genetic lineage tracing and cell depletion experiments in newly generated knock-in Cre/CreERT2 lines, we show here that rare Prrx1-expressing cells act as stem cells for bone, white adipose tissue and dermis in adult mice, which are indispensable for the homeostasis and repair of these tissues. Single-cell profiling reveals the cycling and multipotent nature of Prrx1-expressing cells and the stemness of these cells is further validated by transplantation assays. Moreover, we identify the cell surface markers for Prrx1-expressing stem cells and show that the activities of these stem cells are regulated by Wnt signaling. These findings expand our knowledge of connective tissue homeostasis/regeneration and may help improve stem-cell-based therapies.


Assuntos
Tecido Adiposo Branco , Células-Tronco , Camundongos , Animais
2.
Mol Genet Genomic Med ; 10(10): e2043, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36004822

RESUMO

BACKGROUND: Melorheostosis (MEL) is an exceptionally rare sclerosing bone dysplasia with asymmetrically exuberant bone formation and soft tissue lesions in a segmental distribution. We aimed to summarize the clinical characteristics of Chinese MEL patients and identify their pathogenic cause. METHODS: In total, 10 Chinese MEL patients were recruited, and clinical manifestations and radiological characteristics were recorded. Sanger sequencing of the LEMD3 gene was performed on peripheral blood samples of all patients, while the exome sequencing of matched peripheral blood, melorheostotic bone, and skin lesion samples was conducted on one patient who provided affected bone and skin tissues. Micro-computed tomography (micro-CT) was also used to scan the melorheostotic bone tissue. RESULTS: We found the average age of the 10 MEL patients was 29.5 years (range 11-40 years), and the major symptoms were bone pain, restricted movement, and bone deformity. The lesions sites were mainly located in femur (8/10), tibia (8/10), fibula (6/10), and foot (7/10), the next was pelvis (4/10), and the last were patella (1/10), hand (1/10) and spine (1/10). Radiological examinations showed a mixture of hyperostosis consisting of classic "dripping candle wax," "osteoma-like," or "myositis ossificans-like" patterns in most patients. No germline pathogenic variants in the LEMD3 gene were found in all patients, but a disease-causing somatic variant of MAP2K1 (c.167A > C, p.Gln56Pro) was detected in melorheostotic bone from one patient. Moreover, the micro-CT analysis showed increased porosity in the melorheostotic bone with somatic MAP2K1 variant. CONCLUSION: This is a summary of the clinical characteristics of Chinese MEL patients and we first identify the somatic MAP2K1 variant in Chinese patients. Our findings validate the molecular genetic mechanism of MEL and broaden its phenotype spectrum in the Chinese population.


Assuntos
Melorreostose , Osso e Ossos/patologia , China , Humanos , MAP Quinase Quinase 1/genética , Melorreostose/diagnóstico por imagem , Melorreostose/genética , Melorreostose/patologia , Sequenciamento do Exoma , Microtomografia por Raio-X
3.
Bone Res ; 10(1): 42, 2022 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-35624094

RESUMO

Toll-like receptors (TLRs) play pivotal roles in inflammation and provide important links between the immune and skeletal systems. Although the activation of TLRs may affect osteoclast differentiation and bone metabolism, whether and how TLRs are required for normal bone remodeling remains to be fully explored. In the current study, we show for the first time that TLR9-/- mice exhibit a low bone mass and low-grade systemic chronic inflammation, which is characterized by the expansion of CD4+ T cells and increased levels of inflammatory cytokines, including TNFα, RANKL, and IL1ß. The increased levels of these cytokines significantly promote osteoclastogenesis and induce bone loss. Importantly, TLR9 deletion alters the gut microbiota, and this dysbiosis is the basis of the systemic inflammation and bone loss observed in TLR9-/- mice. Furthermore, through single-cell RNA sequencing, we identified myeloid-biased hematopoiesis in the bone marrow of TLR9-/- mice and determined that the increase in myelopoiesis, likely caused by the adaptation of hematopoietic stem cells to systemic inflammation, also contributes to inflammation-induced osteoclastogenesis and subsequent bone loss in TLR9-/- mice. Thus, our study provides novel evidence that TLR9 signaling connects the gut microbiota, immune system, and bone and is critical in maintaining the homeostasis of inflammation, hematopoiesis, and bone metabolism under normal conditions.

4.
Biomaterials ; 283: 121465, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35286850

RESUMO

Repair of critical-size bone defects in patients with diabetes mellitus (DM) has always been a challenge in clinical treatment. The process of bone defect regeneration can be impaired by underlying diseases including DM, but the mechanism remains unclear. In bone tissue engineering, the integration of bionic coatings and bioactive components into basic scaffolds are common function-enhancing strategies. Small extracellular vesicles (sEVs) have been applied for cell-free tissue regeneration in the last few years. We previously reported that sEVs have flexible and easily-extensible potential, through modular design and engineering modification. The impairment of CD31hiendomucinhi endothelial cells (ECs) whose function is coupling of osteogenesis and angiogenesis, is considered an important contributor to diabetic bone osteopathy, and ZEB1, which is highly expressed in CD31hiendomucinhi ECs, promotes angiogenesis-dependent bone formation. Thus we believe these ECs hold much promise for use in bone regeneration. In addition, c(RGDfC) has been reported to be a highly-effective peptide targeting αvß3, which is highly expressed in the bone microenvironment. In this study, we developed a hyaluronic acid (HA)/poly-L-lysine (PLL) layer-by-layer (LbL) self-assembly coating on ß-TCP (ß-tricalcium phosphate) scaffolds providing immobilization of modularized engineered sEVs (with c(RGDfC) surface functionalization and ZEB1 loading) to facilitate bone defect regeneration under DM conditions. RNA-seq was used to explore possible molecular mechanisms, and the therapeutic effects of bone regeneration were systematically evaluated in vitro and in vivo. Our data demonstrated that this strategy could be very effective in promoting the repair of diabetic bone defects, by enhancing angiogenesis, promoting osteogenesis and inhibiting osteoclast formation.


Assuntos
Diabetes Mellitus , Vesículas Extracelulares , Regeneração Óssea , Fosfatos de Cálcio/química , Diabetes Mellitus/terapia , Células Endoteliais , Humanos , Osteogênese , Engenharia Tecidual , Alicerces Teciduais/química , Homeobox 1 de Ligação a E-box em Dedo de Zinco
5.
Front Cell Dev Biol ; 9: 715042, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34557487

RESUMO

Genetic skeletal dysplasias (GSDs) are a type of disease with complex phenotype and high heterogeneity, characterized by cartilage and bone growth abnormalities. The variable phenotypes of GSD make clinical diagnosis difficult. To explore the clinical utility of targeted exome sequencing (TES) in the diagnosis of GSD, 223 probands with suspected GSD were enrolled for TES with a panel of 322 known disease-causing genes. After bioinformatics analysis, all candidate variants were prioritized by pathogenicity. Sanger sequencing was used to verify candidate variants in the probands and parents and to trace the source of variants in family members. We identified the molecular diagnoses for 110/223 probands from 24 skeletal disorder groups and confirmed 129 pathogenic/likely pathogenic variants in 48 genes. The overall diagnostic rate was 49%. The molecular diagnostic results modified the diagnosis in 25% of the probands, among which mucopolysaccharidosis and spondylo-epi-metaphyseal dysplasias were more likely to be misdiagnosed. The clinical management of 33% of the probands also improved; 21 families received genetic counseling; 4 families accepted prenatal genetic diagnosis, 1 of which was detected to carry pathogenic variants. The results showed that TES achieved a high diagnostic rate for GSD, helping clinicians confirm patients' molecular diagnoses, formulate treatment directions, and carry out genetic counseling. TES could be an economical diagnostic method for patients with GSD.

6.
J Bone Miner Res ; 36(6): 1088-1103, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33599011

RESUMO

Paget's disease of bone (PDB) is a late-onset chronic progressive bone disease characterized by abnormal activation of osteoclasts that results in bone pain, deformities, and fractures. PDB is very rare in Asia. A subset of PDB patients have early onset and can develop malignant giant cell tumors (GCTs) of the bone (PDB/GCTs), which arise within Paget bone lesions; the result is a significantly higher mortality rate. SQSTM1, TNFRSF11A, OPG, VCP, and HNRNPA2B1 have been identified as pathogenic genes of PDB, and ZNF687 is the only confirmed gene to date known to cause PDB/GCT. However, the molecular mechanism underlying PDB/GCT has not been fully elucidated. Here, we investigate an extended Chinese pedigree with eight individuals affected by early-onset and polyostotic PDB, two of whom developed GCTs. We identified a heterozygous 4-bp deletion in the Profilin 1 (PFN1) gene (c.318_321delTGAC) by genetic linkage analysis and exome sequencing for the family. Sanger sequencing revealed another heterozygous 1-bp deletion in PFN1 (c.324_324delG) in a sporadic early-onset PDB/GCT patient, further proving its causative role. Interestingly, a heterozygous missense mutation of PFN1 (c.335 T > C) was identified in another PDB/GCT family, revealing that not only deletion but also missense mutations in PFN1 can cause PDB/GCT. Furthermore, we established a Pfn1-mutated mouse model (C57BL/6J mice) and successfully obtained Pagetic phenotypes in heterozygous mice, verifying loss of function of PFN1 as the cause of PDB/GCT development. In conclusion, our findings reveal mutations in PFN1 as the pathological mechanism in PDB/GCT, and we successfully established Pfn1-mutated mice as a suitable animal model for studying PDB-associated pathological mechanisms. The identification of PFN1 mutations has great diagnostic value for identifying PDB individuals predisposed toward developing GCTs. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Tumores de Células Gigantes , Osteíte Deformante , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Osteíte Deformante/genética , Profilinas/genética , Proteína Sequestossoma-1/genética
7.
Adv Biosyst ; 4(9): e2000152, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32803878

RESUMO

The prognosis for osteosarcoma (OS) continues to be unsatisfactory due to tumor recurrence as a result of metastasis and drug resistance. Several studies have shown that Ewing sarcoma associated transcript 1 (EWSAT1) plays an important role in the progression of OS. Exosomes (Exos) act as important carriers in intercellular communication and play an important role in the tumor microenvironment, especially in tumor-induced angiogenesis. Nonetheless, the specific mechanism via which EWSAT1 and Exos regulate OS progression is unknown, and whether they can be effective therapeutic targets also requires verification. Hence, in this study, it is aimed to investigate the mechanisms of action of EWSAT1 and Exos. EWSAT1 significantly promotes proliferation, migration, colony formation, and survival of OS. EWSAT1 regulates OS-induced angiogenesis via two mechanisms, called the "double stacking effect," which is a combination of the increase in sensitivity/reactivity of vascular endothelial cells triggered by Exos-carrying EWSAT1, and the EWSAT1-induced increase in angiogenic factor secretion. In vivo experiments further validates the "double stacking effect" and shows that EWSAT1-KD effectively inhibits tumor growth in OS. The above observations indicate that EWSAT1 can be used as not only a potential diagnostic and prognostic marker, but also as a precise therapeutic target for OS.


Assuntos
Exossomos/metabolismo , Neovascularização Patológica/metabolismo , Osteossarcoma , RNA Longo não Codificante , Animais , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
8.
J Orthop Surg Res ; 15(1): 303, 2020 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-32762716

RESUMO

BACKGROUND: Statins are the most widely used drugs in elderly patients; the most common clinical application of statins is in aged hyperlipemia patients. There are few studies on the effects and mechanisms of statins on bone in elderly mice with hyperlipemia. The study is to examine the effects of atorvastatin on bone phenotypes and metabolism in aged apolipoprotein E-deficient (apoE-/-) mice, and the possible mechanisms involved in these changes. METHODS: Twenty-four 60-week-old apoE-/- mice were randomly allocated to two groups. Twelve mice were orally gavaged with atorvastatin (10 mg/kg body weight/day) for 12 weeks; the others served as the control group. Bone mass and skeletal microarchitecture were determined using micro-CT. Bone metabolism was assessed by serum analyses, qRT-PCR, and Western blot. Bone marrow-derived mesenchymal stem cells (BMSCs) from apoE-/- mice were differentiated into osteoblasts and treated with atorvastatin and silent information regulator 1 (Sirt1) inhibitor EX-527. RESULTS: The results showed that long-term administration of atorvastatin increases bone mass and improves bone microarchitecture in trabecular bone but not in cortical bone. Furthermore, the serum bone formation marker osteocalcin (OCN) was ameliorated by atorvastatin, whereas the bone resorption marker tartrate-resistant acid phosphatase 5b (Trap5b) did not appear obviously changes after the treatment of atorvastatin. The mRNA expression of Sirt1, runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP), and OCN in bone tissue were increased after atorvastatin administration. Western blot showed same trend in Sirt1 and Runx2. The in vitro study showed that when BMSCs from apoE-/- mice were pretreated with EX527, the higher expression of Runx2, ALP, and OCN activated by atorvastatin decreased significantly or showed no difference compared with the control. The protein expression of Runx2 showed same trend. CONCLUSIONS: Accordingly, the current study validates the hypothesis that atorvastatin can increase bone mass and promote osteogenesis in aged apoE-/- mice by regulating the Sirt1-Runx2 axis.


Assuntos
Atorvastatina/farmacologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteogênese/efeitos dos fármacos , Sirtuína 1/metabolismo , Administração Oral , Idoso , Fosfatase Alcalina/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso Esponjoso/efeitos dos fármacos , Osso Esponjoso/metabolismo , Carbazóis/metabolismo , Carbazóis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/efeitos dos fármacos , Osteocalcina/sangue , Osteocalcina/efeitos dos fármacos , Microtomografia por Raio-X/métodos
9.
Elife ; 82019 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-31482846

RESUMO

Indian Hedgehog (IHH) signaling, a key regulator of skeletal development, is highly activated in cartilage and bone tumors. Yet deletion of Ptch1, encoding an inhibitor of IHH receptor Smoothened (SMO), in chondrocyte or osteoblasts does not cause tumorigenesis. Here, we show that Ptch1 deletion in mice Prrx1+mesenchymal stem/stromal cells (MSCs) promotes MSC proliferation and osteogenic and chondrogenic differentiation but inhibits adipogenic differentiation. Moreover, Ptch1 deletion led to development of osteoarthritis-like phenotypes, exostoses, enchondroma, and osteosarcoma in Smo-Gli1/2-dependent manners. The cartilage and bone tumors are originated from Prrx1+ lineage cells and express low levels of osteoblast and chondrocyte markers, respectively. Mechanistically, Ptch1 deletion increases the expression of Wnt5a/6 and leads to enhanced ß-Catenin activation. Inhibiting Wnt/ß-Catenin pathway suppresses development of skeletal anomalies including enchondroma and osteosarcoma. These findings suggest that cartilage/bone tumors arise from their early progenitor cells and identify the Wnt/ß-Catenin pathway as a pharmacological target for cartilage/bone neoplasms.


Assuntos
Neoplasias Ósseas/fisiopatologia , Proteínas Hedgehog/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Camundongos , Receptor Patched-1/deficiência
10.
J Clin Invest ; 129(5): 1895-1909, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30830877

RESUMO

Bone osteogenic sarcoma has a poor prognosis as the exact cell of origin and the signaling pathways underling tumor formation remain undefined. Here, we report an osteogenic tumor mouse model based on the conditional knockout of liver kinase b1 (Lkb1; also known as Stk11) in Cathepsin K (Ctsk)-Cre expressing cells. Lineage tracing studies demonstrated that Ctsk-Cre could label a population of periosteal cells. The cells functioned as mesenchymal progenitors with regard to markers and functional properties. LKB1 deficiency increased proliferation and osteoblast differentiation of Ctsk+ periosteal cells, while downregulation of mTORC1 activity, using Raptor genetic mouse model or mTORC1 inhibitor treatment, ameliorated tumor progression of Ctsk-Cre Lkb1fllfl mice. Xenograft mouse models, using human osteosarcoma cell lines, also demonstrated that LKB1 deficiency promoted tumor formation, while mTOR inhibition suppressed xenograft tumor growth. In summary, we identified periosteum-derived Ctsk-Cre expressing cells as a cell of origin for osteogenic tumor and suggested the LKB1-mTORC1 pathway as a promising target for treatment of osteogenic tumor.


Assuntos
Neoplasias Ósseas/metabolismo , Deleção de Genes , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Células-Tronco Mesenquimais/citologia , Periósteo/citologia , Proteínas Serina-Treonina Quinases/genética , Sarcoma/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Animais , Diferenciação Celular , Linhagem da Célula , Progressão da Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Transplante de Neoplasias , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogênese , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Microtomografia por Raio-X
11.
J Bone Miner Res ; 34(2): 295-309, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30550637

RESUMO

HB-EGF, a member of the EGF superfamily, plays important roles in development and tissue regeneration. However, its functions in skeletal stem cells and skeleton development and growth remain poorly understood. Here, we used the Cre/LoxP system to ablate or express HB-EGF in Dermo1+ mesenchymal stromal cells and their progenies, including chondrocytes and osteoblast lineage cells, and bone marrow stromal cells (BMSCs). Dermo1-Cre; HB-EGFf/f mice only showed a modest increase in bone mass, whereas Dermo1-HB-EGF mice developed progressive chondrodysplasia, chondroma, osteoarthritis-like joint defects, and loss of bone mass and density, which were alleviated by treatment with EGFR inhibitor AG1478. The cartilage defects were recapitulated in chondrocyte-specific HB-EGF overexpression (Col2-HB-EGF) mice with a lesser severity. Dermo1-HB-EGF mice showed an increase in proliferation but defects in differentiation of chondrocytes and osteoblasts. HB-EGF promoted BMSC proliferation via the Akt1 and Erk pathways but inhibited BMSC differentiation via restraining Smad1/5/8 activation. However, Dermo1-HB-EGF mice showed normal osteoclastogenesis and bone resorption. These results reveal an important function of autocrine or paracrine HB-EGF in mesenchymal stromal cell proliferation and differentiation and suggest that EGF signaling needs to be tightly controlled to maintain bone and articular cartilage integrity. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.


Assuntos
Diferenciação Celular , Proliferação de Células , Condrócitos/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/metabolismo , Esqueleto/crescimento & desenvolvimento , Animais , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Condrócitos/patologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Transgênicos , Osteoblastos/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/farmacologia , Esqueleto/metabolismo , Esqueleto/patologia , Proteínas Smad/genética , Proteínas Smad/metabolismo , Tirfostinas/farmacologia
12.
Cell Discov ; 3: 16050, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28163927

RESUMO

Protein translation regulation has essential roles in inflammatory responses, cancer initiation and the pathogenesis of several neurodegenerative disorders. However, the role of the regulation of protein translation in mammalian skeleton development has been rarely elaborated. Here we report that the lack of the RNA-binding protein sterile alpha motif domain containing protein 4 (SAMD4) resulted in multiple developmental defects in mice, including delayed bone development and decreased osteogenesis. Samd4-deficient mesenchymal progenitors exhibit impaired osteoblast differentiation and function. Mechanism study demonstrates that SAMD4 binds the Mig6 mRNA and inhibits MIG6 protein synthesis. Consistent with this, Samd4-deficient cells have increased MIG6 protein level and knockdown of Mig6 rescues the impaired osteogenesis in Samd4-deficient cells. Furthermore, Samd4-deficient mice also display chondrocyte defects, which is consistent with the regulation of MIG6 protein level by SAMD4. These findings define SAMD4 as a previously unreported key regulator of osteoblastogenesis and bone development, implying that regulation of protein translation is an important mechanism governing skeletogenesis and that control of protein translation could have therapeutic potential in metabolic bone diseases, such as osteoporosis.

13.
J Recept Signal Transduct Res ; 37(3): 217-223, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27485180

RESUMO

CONTEXT: MicroRNA-128 (miR-128), a brain-enriched microRNA, has been reported to participate in the regulation of cell differentiation, but its potential roles in adipogenic and osteogenic differentiation of human mesenchymal stem cells (hMSCs) have not been addressed. OBJECTIVE: The study was conducted to investigate the effects and mechanism of miR-128 on adipogenic and osteogenic differentiation of hMSCs. MATERIALS AND METHODS: Morphology of hMSCs, lipid droplets and calcium nodules were observed and photographed by LSM microscopy. Expression of CCAAT/enhancer binding protein-α (C/EBPα), peroxisome proliferator-activated receptor-γ (PPARγ), miR-128, vascular endothelial growth factor (VEGF), osteocalcin (OCN) and Runt-related transcription factor 2 (RUNX2) was determined by RNA preparation and reverse transcription polymerase chain reaction (RT-PCR), protein expression of VEGF was analyzed by Western blot. RESULTS: It was suggested that miR-128 expression showed a 4.56-fold induction by adipogenic treatment and a 58.8% reduction by osteogenic treatment. Over-expression of miR-128, promoted adipogenic differentiation while inhibited osteogenic differentiation. In contrast, adipocyte formation was inhibited and osteogenesis was enhanced in cells slicing miR-128. Furthermore, over-expression of miR-128 down-regulated VEGF expression in adipogenically and osteogenically differentiated cells. We further identified VEGF as a key regulator in miR-128-induced adipogenic and osteogenic differentiation. Following knockdown of VEGF, the effects of over-expression of miR-128 on adipogenic and osteogenic differentiation of hMSCs were limited. CONCLUSION: It was indicated that miR-128 could regulate adipogenic and osteogenic differentiation of hMSCs significantly through the suppression of VEGF pathway.


Assuntos
Diferenciação Celular/genética , Células-Tronco Mesenquimais , MicroRNAs/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adipócitos/metabolismo , Adipogenia/genética , Proteína alfa Estimuladora de Ligação a CCAAT/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Humanos , MicroRNAs/biossíntese , Osteocalcina/biossíntese , Osteogênese/genética , PPAR gama/biossíntese , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
14.
Gastroenterol Res Pract ; 2012: 808120, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23125851

RESUMO

Purpose. Systematic reviews of case-control and prospective studies showed a positive association between habitual salt intake and gastric cancer. Given new studies published thereafter, we carried out a meta-analysis to assess the association between dietary salt intake and gastric cancer. Methods. Case-control studies and cohort studies published between January 1992 and January 2012 on PubMed and Embase were searched. We quantified associations between salt intake and gastric cancer with meta-analysis. Results. Eleven studies (7 case controls and 4 cohorts) finally were included in the meta-analysis (total population: n = 2076498; events: n = 12039). The combined odds ratio showed significantly positive association between high salt intake and gastric cancer compared with low salt intake (OR = 2.05, 95% CI [1.60, 2.62]; P < 0.00001). In subgroup meta-analysis, findings were slightly different when analyses were restricted to salty food intake (OR = 2.41, 95% CI [2.08, 2.78]; P < 0.00001) as well as in Asia (OR = 1.27 95% CI [1.22, 1.32]; P < 0.00001). There was no evidence that sample size, exposure assessment substantially influenced the estimate of effects. Conclusions. The systemic review supports the hypothesis that dietary salt intake is positively associated with the risk of gastric cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA