Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.512
Filtrar
1.
BMC Musculoskelet Disord ; 25(1): 537, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38997667

RESUMO

BACKGROUND: Human intervertebral disk degeneration (IVDD) is a sophisticated degenerative pathological process. A key cause of IVDD progression is nucleus pulposus cell (NPC) degeneration, which contributes to excessive endoplasmic reticulum stress in the intervertebral disk. However, the mechanisms underlying IVDD and NPC degeneration remain unclear. METHODS: We used interleukin (IL)-1ß stimulation to establish an NPC-degenerated IVDD model and investigated whether human urine-derived stem cell (USC) exosomes could prevent IL-1ß-induced NPC degeneration using western blotting, quantitative real-time polymerase chain reaction, flow cytometry, and transcriptome sequencing techniques. RESULTS: We successfully extracted and identified USCs and exosomes from human urine. IL-1ß substantially downregulated NPC viability and induced NPC degeneration while modulating the expression of SOX-9, collagen II, and aggrecan. Exosomes from USCs could rescue IL-1ß-induced NPC degeneration and restore the expression levels of SOX-9, collagen II, and aggrecan. CONCLUSIONS: USC-derived exosomes can prevent NPCs from degeneration following IL-1ß stimulation. This finding can aid the development of a potential treatment strategy for IVDD.


Assuntos
Exossomos , Interleucina-1beta , Degeneração do Disco Intervertebral , Núcleo Pulposo , Fatores de Transcrição SOX9 , Humanos , Interleucina-1beta/metabolismo , Exossomos/metabolismo , Degeneração do Disco Intervertebral/patologia , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/terapia , Núcleo Pulposo/metabolismo , Núcleo Pulposo/patologia , Núcleo Pulposo/citologia , Núcleo Pulposo/efeitos dos fármacos , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Animais , Células-Tronco/metabolismo , Células Cultivadas , Agrecanas/metabolismo , Agrecanas/genética , Masculino , Urina/citologia , Urina/química , Feminino , Colágeno Tipo II/metabolismo
2.
Clin Transl Sci ; 17(7): e13881, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38982592

RESUMO

Chondrocyte apoptosis is recognized as one of the pathological features involved in cartilage degeneration driving the onset and progression of knee osteoarthritis (OA). This study aimed to determine the molecular mechanism underlying the effect of clusterin (CLU), anti-apoptotic molecule, in human knee OA chondrocytes. Primary knee OA chondrocytes were isolated from the cartilage of knee OA patients and divided into five groups: (1) the cells treated with interleukin (IL)-1ß, (2) CLU alone, (3) a combination of IL-1ß and CLU, (4) LY294002 (PI3K inhibitor) along with IL-1ß and CLU, and (5) the untreated cells. Production of apoptotic, inflammatory, anabolic, and catabolic mediators in knee OA chondrocytes was determined after treatment for 24 h. Our in vitro study uncovered that CLU significantly suppressed the production of inflammatory mediators [nitric oxide (NO), IL6, and tumor necrosis factor (TNF)-α] and apoptotic molecule (caspase-3, CASP3). CLU significantly upregulated messenger ribonucleic acid (mRNA) expressions of anabolic factors [SRY-box transcription factor-9 (SOX9) and aggrecan (ACAN)], but significantly downregulated mRNA expressions of IL6, nuclear factor kappa-B (NF-κB), CASP3, and matrix metalloproteinase-13 (MMP13). Anti-apoptotic and anti-inflammatory effects of CLU were mediated through activating PI3K/Akt signaling pathway. The findings suggest that CLU might have beneficial effects on knee OA chondrocytes by exerting anti-apoptotic and anti-inflammatory functions via PI3K/Akt pathway, making CLU a promising target for potential therapeutic interventions in knee OA.


Assuntos
Apoptose , Condrócitos , Clusterina , Interleucina-1beta , Osteoartrite do Joelho , Humanos , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Osteoartrite do Joelho/patologia , Osteoartrite do Joelho/metabolismo , Apoptose/efeitos dos fármacos , Clusterina/metabolismo , Clusterina/genética , Interleucina-1beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Cultivadas , Masculino , Pessoa de Meia-Idade , Idoso , Inflamação/metabolismo , Inflamação/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Feminino , Fosfatidilinositol 3-Quinases/metabolismo , Morfolinas/farmacologia , Cromonas/farmacologia , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Metaloproteinase 13 da Matriz/metabolismo , Mediadores da Inflamação/metabolismo , Óxido Nítrico/metabolismo
3.
BMC Cancer ; 24(1): 883, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39039505

RESUMO

BACKGROUND: The role of miR-145-5p in non-small cell lung cancer (NSCLC) has been studied, however, the regulation of hBMSCs-derived exosomes (Exo) transmitted miR-145-5p in NSCLC was still unknown. This study aimed to investigate the role of hBMSCs-derived exosomes (Exo) in the progression of NSCLC. METHODS: The Exo was extracted from hBMSCs and added to A549 and H1299 cell culture, followed by the detection of cell proliferation, migration, and invasion. The correlation between the expression of miR-145-5p and SOX9, as well as their binding relationship was determined by correlation analysis, luciferase gene reporter assay and RNA pull-down assays. The in vivo animal model was established to further verify the impact of hBMSCs-Exo. RESULTS: It showed that miR-145-5p was downregulated and SOX9 was upregulated in NSCLC tissues. HBMSCs-derived Exo, and hBMSCs-Exo with overexpression of miR-145-5p could inhibit cell proliferation, migration, and invasion of both A549 and H1299 cells, and prevent against tumor progression in vivo. MiR-145-5p and SOX9 were found to be able to bind to each other, and a negative correlation were observed between the expression of them in NSCLC tissues. Furthermore, inhibition of SOX9 could reversed the suppressed role of miR-145-5p in vitro and in vivo. CONCLUSION: Therefore, HBMSCs-Exo effectively transmitted miR-145-5p, leading to the suppression of malignant development in NSCLC through the regulation of SOX9.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Movimento Celular , Proliferação de Células , Exossomos , Neoplasias Pulmonares , Células-Tronco Mesenquimais , MicroRNAs , Fatores de Transcrição SOX9 , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Animais , Exossomos/metabolismo , Exossomos/genética , Camundongos , Proliferação de Células/genética , Células-Tronco Mesenquimais/metabolismo , Movimento Celular/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Masculino , Feminino , Células A549 , Ensaios Antitumorais Modelo de Xenoenxerto
4.
BMC Vet Res ; 20(1): 326, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39026327

RESUMO

To evaluate Tribulus terrestris and Mucuna pruriens for inducing all-male tilapia, mixed-sex Nile tilapia, Oreochromis niloticus, (mean weight 0.025 ± 0.009 g; mean length 1.25 ± 0.012 cm), were given a meal supplemented with either T. terrestris powder (commercial fish feed, 40% crude protein) (TT group), M. pruriens seed extract (MP group), MP + TT (mixed group), 17α-methyl testosterone (MT, control positive), or without supplements (control negative). The MP extracts significantly increased (P < 0.05) the final weight, weight gain, weight gain rate, and specific growth rate while feed conversion ratio was significantly decreased (P < 0.05). Plant extracts markedly improved (P < 0.05) the survival rate, proportion of males, and total testosterone compared to control and MT. Estrogen levels were lower in groups with plant extract than other groups. Fifteen days post-feeding, the Amh gene was expressed in the brain of O. niloticus fries with higher levels in MP, TT, and MT groups. Additionally, the expression of the Sox9 and Dmrt1 genes as a male related genes in fish fry gonads revealed significantly (P < 0.05) higher levels in groups fed on MP, TT, and MT compared to control after 30-day post-feeding, whereas; Foxl2 gene expression as a female related gene was significantly (P < 0.05) lower in fish fed on MP, TT, and MT compared to other groups after 30 days post feeding. Histologically, MT, MP, TT, and the mixture all exhibited solely male reproductive traits without noticeable abnormalities. This study concluded that each of the TT or MP extracts can induce sex reversal in tilapia while having no negative health impact compared to MT as the growth and survival rate in the treated groups with TT and MP were higher than control and group treated with MT.


Assuntos
Ração Animal , Ciclídeos , Suplementos Nutricionais , Metiltestosterona , Mucuna , Tribulus , Animais , Masculino , Tribulus/química , Metiltestosterona/farmacologia , Ração Animal/análise , Mucuna/química , Ciclídeos/crescimento & desenvolvimento , Ciclídeos/genética , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Hormônio Antimülleriano/genética , Hormônio Antimülleriano/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Dieta/veterinária , Proteína Forkhead Box L2/genética , Feminino , Testosterona/sangue
5.
Eur J Cell Biol ; 103(2): 151424, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38823166

RESUMO

Actin is a central mediator of the chondrocyte phenotype. Monolayer expansion of articular chondrocytes on tissue culture polystyrene, for cell-based repair therapies, leads to chondrocyte dedifferentiation. During dedifferentiation, chondrocytes spread and filamentous (F-)actin reorganizes from a cortical to a stress fiber arrangement causing a reduction in cartilage matrix expression and an increase in fibroblastic matrix and contractile molecule expression. While the downstream mechanisms regulating chondrocyte molecular expression by alterations in F-actin organization have become elucidated, the critical upstream regulators of F-actin networks in chondrocytes are not completely known. Tropomyosin (TPM) and the RhoGTPases are known regulators of F-actin networks. The main purpose of this study is to elucidate the regulation of passaged chondrocyte F-actin stress fiber networks and cell phenotype by the specific TPM, TPM3.1, and the RhoGTPase, CDC42. Our results demonstrated that TPM3.1 associates with cortical F-actin and stress fiber F-actin in primary and passaged chondrocytes, respectively. In passaged cells, we found that pharmacological TPM3.1 inhibition or siRNA knockdown causes F-actin reorganization from stress fibers back to cortical F-actin and causes an increase in G/F-actin. CDC42 inhibition also causes formation of cortical F-actin. However, pharmacological CDC42 inhibition, but not TPM3.1 inhibition, leads to the re-association of TPM3.1 with cortical F-actin. Both TPM3.1 and CDC42 inhibition, as well as TPM3.1 knockdown, reduces nuclear localization of myocardin related transcription factor, which suppresses dedifferentiated molecule expression. We confirmed that TPM3.1 or CDC42 inhibition partially redifferentiates passaged cells by reducing fibroblast matrix and contractile expression, and increasing chondrogenic SOX9 expression. A further understanding on the regulation of F-actin in passaged cells may lead into new insights to stimulate cartilage matrix expression in cells for regenerative therapies.


Assuntos
Actinas , Desdiferenciação Celular , Condrócitos , Fibras de Estresse , Tropomiosina , Condrócitos/metabolismo , Condrócitos/citologia , Fibras de Estresse/metabolismo , Animais , Actinas/metabolismo , Tropomiosina/metabolismo , Tropomiosina/genética , Fenótipo , Células Cultivadas , Proteína cdc42 de Ligação ao GTP/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Transativadores/metabolismo , Transativadores/genética
6.
JCI Insight ; 9(11)2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38855864

RESUMO

The transcription factor SRY-related HMG box 9 (Sox9) is essential for chondrogenesis. Mutations in and around SOX9 cause campomelic dysplasia (CD) characterized by skeletal malformations. Although the function of Sox9 in this context is well studied, the mechanisms that regulate Sox9 expression in chondrocytes remain to be elucidated. Here, we have used genome-wide profiling to identify 2 Sox9 enhancers located in a proximal breakpoint cluster responsible for CD. Enhancer activity of E308 (located 308 kb 5' upstream) and E160 (located 160 kb 5' upstream) correlated with Sox9 expression levels, and both enhancers showed a synergistic effect in vitro. While single deletions in mice had no apparent effect, simultaneous deletion of both E308 and E160 caused a dwarf phenotype, concomitant with a reduction of Sox9 expression in chondrocytes. Moreover, bone morphogenetic protein 2-dependent chondrocyte differentiation of limb bud mesenchymal cells was severely attenuated in E308/E160 deletion mice. Finally, we found that an open chromatin region upstream of the Sox9 gene was reorganized in the E308/E160 deletion mice to partially compensate for the loss of E308 and E160. In conclusion, our findings reveal a mechanism of Sox9 gene regulation in chondrocytes that might aid in our understanding of the pathophysiology of skeletal disorders.


Assuntos
Displasia Campomélica , Diferenciação Celular , Condrócitos , Condrogênese , Fatores de Transcrição SOX9 , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Animais , Condrócitos/metabolismo , Camundongos , Displasia Campomélica/genética , Displasia Campomélica/patologia , Displasia Campomélica/metabolismo , Condrogênese/genética , Diferenciação Celular/genética , Elementos Facilitadores Genéticos/genética , Cromatina/metabolismo , Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos Knockout , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 2/genética , Humanos , Desenvolvimento Ósseo/genética
7.
Exp Cell Res ; 440(2): 114135, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38901791

RESUMO

Liver fibrosis is a significant health concern globally due to its association with severe liver conditions like cirrhosis and liver cancer. Histone lactylation has been implicated in the progression of hepatic fibrosis, but its specific role in liver fibrosis, particularly regarding H3K18 lactylation, remained unclear. To investigate this, we established in vivo and in vitro models of liver fibrosis using carbon tetrachloride (CCl4) injection in rats and stimulation of hepatic stellate cells (HSCs) with TGF-ß1, respectively. We found that histone lactylation, particularly H3K18 lactylation, was upregulated in both CCl4-induced rats and TGF-ß1-activated HSCs, indicating its potential involvement in liver fibrosis. Further experiments revealed that lactate dehydrogenase A (LDHA) knockdown inhibited H3K18 lactylation and had a beneficial effect on liver fibrosis by suppressing HSC proliferation, migration, and extracellular matrix (ECM) deposition. This suggests that H3K18 lactylation promotes liver fibrosis progression. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays demonstrated that H3K18 lactylation facilitated the transcription of SOX9, a transcription factor associated with fibrosis. Importantly, overexpression of SOX9 counteracted the effects of LDHA silencing on activated HSCs, indicating that SOX9 is downstream of H3K18 lactylation in promoting liver fibrosis. In summary, this study uncovers a novel mechanism by which H3K18 lactylation contributes to liver fibrosis by activating SOX9 transcription. This finding opens avenues for exploring new therapeutic strategies for hepatic fibrosis targeting histone lactylation pathways.


Assuntos
Progressão da Doença , Células Estreladas do Fígado , Histonas , Cirrose Hepática , Ratos Sprague-Dawley , Fatores de Transcrição SOX9 , Animais , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/genética , Cirrose Hepática/induzido quimicamente , Histonas/metabolismo , Histonas/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Ratos , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Masculino , Proliferação de Células , Tetracloreto de Carbono , Fator de Crescimento Transformador beta1/metabolismo , Humanos , Transcrição Gênica , Movimento Celular/genética , L-Lactato Desidrogenase/metabolismo , L-Lactato Desidrogenase/genética
8.
Osteoarthritis Cartilage ; 32(8): 872-885, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38852879

RESUMO

OBJECTIVE: Understanding the mechanisms of hip disease, such as osteoarthritis (OA), is crucial to advance their treatment. Such hip diseases often involve specific morphological changes. Genetic variations, called single nucleotide polymorphisms (SNPs), influence various hip morphological parameters. This study investigated the biological relevance of SNPs correlated to hip morphology in genome-wide association studies (GWAS). The SNP-associated genes were compared to genes associated with OA in other joints, aiming to see if the same genes play a role in both hip development and the risk of OA in other lower limb joints. METHODOLOGY: A systematic literature review was conducted to identify SNPs correlated with hip morphology, based on the Population, Intervention, Comparison, Outcome, and Study (PICOS) framework. Afterwards, Gene Ontology (GO) analysis was performed, using EnrichR, on the SNP-associated genes and compared with non-hip OA-associated genes, across different databases. RESULTS: Reviewing 49 GWAS identified 436 SNPs associated with hip joint morphology, encompassing variance in bone size, structure and shape. Among the SNP-associated genes, SOX9 plays a pivotal role in size, GDF5 impacts bone structure, and BMP7 affects shape. Overall, skeletal system development, regulation of cell differentiation, and chondrocyte differentiation emerged as crucial processes influencing hip morphology. Eighteen percent of GWAS-identified genes related to hip morphology were also associated with non-hip OA. CONCLUSION: Our findings indicate the existence of multiple shared genetic mechanisms across hip morphology and OA, highlighting the necessity for more extensive research in this area, as in contrast to the hip, the genetic background on knee or foot morphology remains largely understudied.


Assuntos
Estudo de Associação Genômica Ampla , Fator 5 de Diferenciação de Crescimento , Articulação do Quadril , Osteoartrite do Quadril , Polimorfismo de Nucleotídeo Único , Humanos , Osteoartrite do Quadril/genética , Osteoartrite do Quadril/patologia , Fator 5 de Diferenciação de Crescimento/genética , Articulação do Quadril/patologia , Proteína Morfogenética Óssea 7/genética , Fatores de Transcrição SOX9/genética , Predisposição Genética para Doença
9.
Sci Rep ; 14(1): 12297, 2024 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811798

RESUMO

The current study aimed to investigate the effect of Sox9-Cre-directed Nr5a1-conditional knockout (Sox9-Cre;Nr5a1flox/flox) on adrenal development. We showed that SOX9 is expressed by adrenocortical cells at E10.5-E11.5 but is extinguished no later than E12.5. The number of adrenocortical cells significantly reduced in Sox9-Cre;Nr5a1flox/flox mice while the number of cleaved caspase 3-positive cells increased compared to that in the controls at E11.5-E12.5, when the adrenal primordium (AP) is about to expand. This indicated that fetal adrenocortical cells are lost via apoptosis due to Nr5a1 ablation by E12.5. Both medulla formation and encapsulation were perturbed, accompanied by a smaller AP size, in Sox9-Cre;Nr5a1flox/flox mice during embryonic development. Adult Sox9-Cre;Nr5a1flox/flox adrenals were hypoplastic and exhibited irregular organization of the medulla with aberrant sex differentiation in the X zone. Additionally, there were histologically eosin-negative vacuolated cells, which were negative for both the X-zone marker 20αHSD and the steroidogenesis marker 3ßHSD at the innermost cortex of Sox9-Cre;Nr5a1flox/flox adrenals. Although Nr5a1+/- adrenals were hypoplastic, a small number of chromaffin cells were properly located in the center, having normal sex differences in the X-zone. The results collectively provided in-vivo evidence that Nr5a1 plays a critical role in AP expansion and subsequent adrenal development.


Assuntos
Glândulas Suprarrenais , Fatores de Transcrição SOX9 , Fator Esteroidogênico 1 , Animais , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Camundongos , Fator Esteroidogênico 1/metabolismo , Fator Esteroidogênico 1/genética , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/embriologia , Integrases/metabolismo , Integrases/genética , Camundongos Knockout , Feminino , Masculino
10.
J Oral Rehabil ; 51(8): 1507-1520, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38717032

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) derived from the synovium, known as synovium mesenchymal stem cells (SMSCs), exhibit significant potential for articular cartilage regeneration owing to their capacity for chondrogenic differentiation. However, the microRNAs (miRNAs) governing this process and the associated mechanisms remain unclear. While mechanical stress positively influences chondrogenesis in MSCs, the miRNA-mediated response of SMSCs to mechanical stimuli is not well understood. OBJECTIVE: This study explores the miRNA-driven mechano-transduction in SMSCs chondrogenesis under mechanical stress. METHODS: The surface phenotype of SMSCs was analysed by flow cytometry. Chondrogenesis capacities of SMSCs were examined by Alcian blue staining. High throughput sequencing was used to screen mechano-sensitive miRNAs of SMSCs. The RNA expression level of COL2A1, ACAN, SOX9, BMPR2 and miR-143-3p of SMSCs were tested by quantitative real-time polymerase chain reaction (qRT-PCR). The interaction between miR-143-3p and TLR4 was confirmed by luciferase reporter assays. The protein expression levels of related genes were assessed by western blot. RESULTS: High-throughput sequencing revealed a notable reduction in miR-143-3p levels in mechanically stressed SMSCs. Gain- or loss-of-function strategies introduced by lentivirus demonstrated that miR-143-3p overexpression hindered chondrogenic differentiation, whereas its knockdown promoted this process. Bioinformatics scrutiny and luciferase reporter assays pinpointed a potential binding site for miR-143-3p within the 3'-UTR of bone morphogenetic protein receptor type 2 (BMPR2). MiR-143-3p overexpression decreased BMPR2 expression and phosphorylated Smad1, 5 and 8 levels, while its inhibition activated BMPR2-Smad pathway. CONCLUSION: This study elucidated that miR-143-3p negatively regulates SMSCs chondrogenic differentiation through the BMPR2-Smad pathway under mechanical tensile stress. The direct targeting of BMPR2 by miR-143-3p established a novel dimension to our understanding of mechano-transduction mechanism during SMSC chondrogenesis. This understanding is crucial for advancing strategies in articular cartilage regeneration.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II , Diferenciação Celular , Condrogênese , Células-Tronco Mesenquimais , MicroRNAs , Transdução de Sinais , Estresse Mecânico , Membrana Sinovial , Humanos , Agrecanas/metabolismo , Agrecanas/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Condrogênese/fisiologia , Colágeno Tipo II/metabolismo , Colágeno Tipo II/genética , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , MicroRNAs/genética , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo
11.
Cancer Lett ; 593: 216939, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38729556

RESUMO

Helicobacter pylori (H. pylori) infection is the main risk factor for gastric cancer. The SRY-Box Transcription Factor 9 (SOX9) serves as a marker of stomach stem cells. We detected strong associations between AURKA and SOX9 expression levels in gastric cancers. Utilizing in vitro and in vivo mouse models, we demonstrated that H. pylori infection induced elevated levels of both AURKA and SOX9 proteins. Notably, the SOX9 protein and transcription activity levels were dependent on AURKA expression. AURKA knockdown led to a reduction in the number and size of gastric gland organoids. Conditional knockout of AURKA in mice resulted in a decrease in SOX9 baseline level in AURKA-knockout gastric glands, accompanied by diminished SOX9 induction following H. pylori infection. We found an AURKA-dependent increase in EIF4E and cap-dependent translation with an AURKA-EIF4E-dependent increase in SOX9 polysomal RNA levels. Immunoprecipitation assays demonstrated binding of AURKA to EIF4E with a decrease in EIF4E ubiquitination. Immunohistochemistry analysis on tissue arrays revealed moderate to strong immunostaining of AURKA and SOX9 with a significant correlation in gastric cancer tissues. These findings elucidate the mechanistic role of AURKA in regulating SOX9 levels via cap-dependent translation in response to H. pylori infection in gastric tumorigenesis.


Assuntos
Aurora Quinase A , Fator de Iniciação 4E em Eucariotos , Infecções por Helicobacter , Helicobacter pylori , Fatores de Transcrição SOX9 , Neoplasias Gástricas , Animais , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Humanos , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Aurora Quinase A/metabolismo , Aurora Quinase A/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Fator de Iniciação 4E em Eucariotos/genética , Camundongos Knockout , Camundongos , Biossíntese de Proteínas , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Ubiquitinação
12.
Int J Biol Sci ; 20(7): 2686-2697, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38725852

RESUMO

Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer. Breast cancer stem cells (BCSCs) are believed to play a crucial role in the carcinogenesis, therapy resistance, and metastasis of TNBC. It is well known that inflammation promotes stemness. Several studies have identified breast cancer-associated gene 2 (BCA2) as a potential risk factor for breast cancer incidence and prognosis. However, whether and how BCA2 promotes BCSCs has not been elucidated. Here, we demonstrated that BCA2 specifically promotes lipopolysaccharide (LPS)-induced BCSCs through LPS induced SOX9 expression. BCA2 enhances the interaction between myeloid differentiation primary response protein 88 (MyD88) and Toll-like receptor 4 (TLR4) and inhibits the interaction of MyD88 with deubiquitinase OTUD4 in the LPS-mediated NF-κB signaling pathway. And SOX9, an NF-κB target gene, mediates BCA2's pro-stemness function in TNBC. Our findings provide new insights into the molecular mechanisms by which BCA2 promotes breast cancer and potential therapeutic targets for the treatment of breast cancer.


Assuntos
Neoplasias da Mama , Células-Tronco Neoplásicas , Fatores de Transcrição SOX9 , Ubiquitina-Proteína Ligases , Feminino , Humanos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Células-Tronco Neoplásicas/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Regulação para Cima
13.
Differentiation ; 138: 100782, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38810379

RESUMO

The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.


Assuntos
Mandíbula , Crista Neural , Animais , Embrião de Galinha , Camundongos , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Padronização Corporal/genética , Cartilagem/metabolismo , Cartilagem/crescimento & desenvolvimento , Cartilagem/citologia , Diferenciação Celular , Galinhas/genética , Cílios/metabolismo , Cílios/genética , Regulação da Expressão Gênica no Desenvolvimento , Mandíbula/crescimento & desenvolvimento , Mandíbula/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Mesoderma/crescimento & desenvolvimento , Crista Neural/citologia , Crista Neural/metabolismo , Transdução de Sinais , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética
14.
BMC Genomics ; 25(1): 464, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38741085

RESUMO

Gonad development includes sex determination and divergent maturation of the testes and ovaries. Recent advances in measuring gene expression in single cells are providing new insights into this complex process. However, the underlying epigenetic regulatory mechanisms remain unclear. Here, we profiled chromatin accessibility in mouse gonadal cells of both sexes from embryonic day 11.5 to 14.5 using single-cell assay for transposase accessible chromatin by sequencing (scATAC-seq). Our results showed that individual cell types can be inferred by the chromatin landscape, and that cells can be temporally ordered along developmental trajectories. Integrative analysis of transcriptomic and chromatin-accessibility maps identified multiple putative regulatory elements proximal to key gonadal genes Nr5a1, Sox9 and Wt1. We also uncover cell type-specific regulatory factors underlying cell type specification. Overall, our results provide a better understanding of the epigenetic landscape associated with the progressive restriction of cell fates in the gonad.


Assuntos
Linhagem da Célula , Cromatina , Gônadas , Fatores de Transcrição SOX9 , Análise de Célula Única , Animais , Cromatina/metabolismo , Cromatina/genética , Camundongos , Linhagem da Célula/genética , Feminino , Masculino , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Gônadas/metabolismo , Gônadas/citologia , Gônadas/embriologia , Fator Esteroidogênico 1/genética , Fator Esteroidogênico 1/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo , Testículo/metabolismo , Testículo/citologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Ovário/metabolismo , Ovário/citologia
15.
Nat Commun ; 15(1): 4148, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755149

RESUMO

Cell plasticity theoretically extends to all possible cell types, but naturally decreases as cells differentiate, whereas injury-repair re-engages the developmental plasticity. Here we show that the lung alveolar type 2 (AT2)-specific transcription factor (TF), CEBPA, restricts AT2 cell plasticity in the mouse lung. AT2 cells undergo transcriptional and epigenetic maturation postnatally. Without CEBPA, both neonatal and mature AT2 cells reduce the AT2 program, but only the former reactivate the SOX9 progenitor program. Sendai virus infection bestows mature AT2 cells with neonatal plasticity where Cebpa mutant, but not wild type, AT2 cells express SOX9, as well as more readily proliferate and form KRT8/CLDN4+ transitional cells. CEBPA promotes the AT2 program by recruiting the lung lineage TF NKX2-1. The temporal change in CEBPA-dependent plasticity reflects AT2 cell developmental history. The ontogeny of AT2 cell plasticity and its transcriptional and epigenetic mechanisms have implications in lung regeneration and cancer.


Assuntos
Células Epiteliais Alveolares , Plasticidade Celular , Fator Nuclear 1 de Tireoide , Animais , Camundongos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/citologia , Fator Nuclear 1 de Tireoide/metabolismo , Fator Nuclear 1 de Tireoide/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Proteínas Estimuladoras de Ligação a CCAAT/genética , Diferenciação Celular , Epigênese Genética , Camundongos Endogâmicos C57BL , Lesão Pulmonar/patologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/genética , Regeneração , Vírus Sendai/genética , Vírus Sendai/fisiologia , Proliferação de Células , Camundongos Knockout , Pulmão/metabolismo
16.
Int J Mol Sci ; 25(10)2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38791601

RESUMO

Osteoarthritis (OA) is a common joint disorder characterized by cartilage degeneration, often leading to pain and functional impairment. Minced cartilage implantation (MCI) has emerged as a promising one-step alternative for large cartilage defects. However, the source of chondrocytes for MCI remains a challenge, particularly in advanced OA, as normal cartilage is scarce. We performed in vitro studies to evaluate the feasibility of MCI using osteophyte cartilage, which is present in patients with advanced OA. Osteophyte and articular cartilage samples were obtained from 22 patients who underwent total knee arthroplasty. Chondrocyte migration and proliferation were assessed using cartilage fragment/atelocollagen composites to compare the characteristics and regenerative potential of osteophytes and articular cartilage. Histological analysis revealed differences in cartilage composition between osteophytes and articular cartilage, with higher expression of type X collagen and increased chondrocyte proliferation in the osteophyte cartilage. Gene expression analysis identified distinct gene expression profiles between osteophytes and articular cartilage; the expression levels of COL2A1, ACAN, and SOX9 were not significantly different. Chondrocytes derived from osteophyte cartilage exhibit enhanced proliferation, and glycosaminoglycan production is increased in both osteophytes and articular cartilage. Osteophyte cartilage may serve as a viable alternative source of MCI for treating large cartilage defects in OA.


Assuntos
Cartilagem Articular , Proliferação de Células , Condrócitos , Osteoartrite , Osteófito , Humanos , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Cartilagem Articular/cirurgia , Condrócitos/metabolismo , Condrócitos/patologia , Osteófito/metabolismo , Osteófito/patologia , Masculino , Feminino , Idoso , Osteoartrite/metabolismo , Osteoartrite/patologia , Osteoartrite/cirurgia , Pessoa de Meia-Idade , Colágeno Tipo II/metabolismo , Colágeno Tipo II/genética , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Células Cultivadas , Movimento Celular
17.
Commun Biol ; 7(1): 545, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714724

RESUMO

CircRNAs are covalently closed, single-stranded RNA that form continuous loops and play a crucial role in the initiation and progression of tumors. Cancer stem cells (CSCs) are indispensable for cancer development; however, the regulation of cancer stem cell-like properties in gastric cancer (GC) and its specific mechanism remain poorly understood. We elucidate the specific role of Circ-0075305 in GC stem cell properties. Circ-0075305 associated with chemotherapy resistance was identified by sequencing GC cells. Subsequent confirmation in both GC tissues and cell lines revealed that patients with high expression of Circ-0075305 had significantly better overall survival (OS) rates than those with low expression, particularly when treated with postoperative adjuvant chemotherapy for GC. In vitro and in vivo experiments confirmed that overexpression of Circ-0075305 can effectively reduce stem cell-like properties and enhance the sensitivity of GC cells to Oxaliplatin compared with the control group. Circ-0075305 promotes RPRD1A expression by acting as a sponge for corresponding miRNAs. The addition of LF3 (a ß-catenin/TCF4 interaction antagonist) confirmed that RPRD1A inhibited the formation of the TCF4-ß-catenin transcription complex through competitive to ß-catenin and suppressed the transcriptional activity of stem cell markers such as SOX9 via the Wnt/ß-catenin signaling pathway. This leads to the downregulation of stem cell-like property-related markers in GC. This study revealed the underlying mechanisms that regulate Circ-0075305 in GCSCs and suggests that its role in reducing ß-catenin signaling may serve as a potential therapeutic candidate.


Assuntos
Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas , RNA Circular , Fatores de Transcrição SOX9 , Neoplasias Gástricas , Fator de Transcrição 4 , beta Catenina , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Humanos , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , beta Catenina/metabolismo , beta Catenina/genética , RNA Circular/genética , RNA Circular/metabolismo , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo , Animais , Camundongos , Linhagem Celular Tumoral , Camundongos Nus , Masculino , Feminino , Resistencia a Medicamentos Antineoplásicos/genética , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade
18.
PLoS One ; 19(5): e0301080, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38728328

RESUMO

Entheses are classified into three types: fibrocartilaginous, fibrous, and periosteal insertions. However, the mechanism behind the development of fibrous entheses and periosteal insertions remains unclear. Since both entheses are part of the temporomandibular joint (TMJ), this study analyzes the TMJ entheses. Here, we show that SOX9 expression is negatively regulated during TMJ enthesis development, unlike fibrocartilage entheses which are modularly formed by SCX and SOX9 positive progenitors. The TMJ entheses was adjacent to the intramembranous bone rather than cartilage. SOX9 expression was diminished during TMJ enthesis development. To clarify the functional role of Sox9 in the development of TMJ entheses, we examined these structures in TMJ using Wnt1Cre;Sox9flox/+ reporter mice. Wnt1Cre;Sox9flox/+ mice showed enthesial deformation at the TMJ. Next, we also observed a diminished SOX9 expression area at the enthesis in contact with the clavicle's membranous bone portion, similar to the TMJ entheses. Together, these findings reveal that the timing of SOX9 expression varies with the ossification development mode.


Assuntos
Osteogênese , Fatores de Transcrição SOX9 , Articulação Temporomandibular , Fatores de Transcrição SOX9/metabolismo , Fatores de Transcrição SOX9/genética , Animais , Camundongos , Articulação Temporomandibular/metabolismo , Articulação Temporomandibular/crescimento & desenvolvimento , Osteogênese/genética , Regulação para Baixo , Fibrocartilagem/metabolismo , Camundongos Transgênicos
19.
J Clin Invest ; 134(11)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38687617

RESUMO

One critical mechanism through which prostate cancer (PCa) adapts to treatments targeting androgen receptor (AR) signaling is the emergence of ligand-binding domain-truncated and constitutively active AR splice variants, particularly AR-V7. While AR-V7 has been intensively studied, its ability to activate distinct biological functions compared with the full-length AR (AR-FL), and its role in regulating the metastatic progression of castration-resistant PCa (CRPC), remain unclear. Our study found that, under castrated conditions, AR-V7 strongly induced osteoblastic bone lesions, a response not observed with AR-FL overexpression. Through combined ChIP-seq, ATAC-seq, and RNA-seq analyses, we demonstrated that AR-V7 uniquely accesses the androgen-responsive elements in compact chromatin regions, activating a distinct transcription program. This program was highly enriched for genes involved in epithelial-mesenchymal transition and metastasis. Notably, we discovered that SOX9, a critical metastasis driver gene, was a direct target and downstream effector of AR-V7. Its protein expression was dramatically upregulated in AR-V7-induced bone lesions. Moreover, we found that Ser81 phosphorylation enhanced AR-V7's pro-metastasis function by selectively altering its specific transcription program. Blocking this phosphorylation with CDK9 inhibitors impaired the AR-V7-mediated metastasis program. Overall, our study has provided molecular insights into the role of AR splice variants in driving the metastatic progression of CRPC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias de Próstata Resistentes à Castração , Receptores Androgênicos , Animais , Humanos , Masculino , Camundongos , Processamento Alternativo , Neoplasias Ósseas/secundário , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Metástase Neoplásica , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/patologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Transcrição Gênica
20.
Signal Transduct Target Ther ; 9(1): 96, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38653754

RESUMO

The translocation of YAP from the cytoplasm to the nucleus is critical for its activation and plays a key role in tumor progression. However, the precise molecular mechanisms governing the nuclear import of YAP are not fully understood. In this study, we have uncovered a crucial role of SOX9 in the activation of YAP. SOX9 promotes the nuclear translocation of YAP by direct interaction. Importantly, we have identified that the binding between Asp-125 of SOX9 and Arg-124 of YAP is essential for SOX9-YAP interaction and subsequent nuclear entry of YAP. Additionally, we have discovered a novel asymmetrical dimethylation of YAP at Arg-124 (YAP-R124me2a) catalyzed by PRMT1. YAP-R124me2a enhances the interaction between YAP and SOX9 and is associated with poor prognosis in multiple cancers. Furthermore, we disrupted the interaction between SOX9 and YAP using a competitive peptide, S-A1, which mimics an α-helix of SOX9 containing Asp-125. S-A1 significantly inhibits YAP nuclear translocation and effectively suppresses tumor growth. This study provides the first evidence of SOX9 as a pivotal regulator driving YAP nuclear translocation and presents a potential therapeutic strategy for YAP-driven human cancers by targeting SOX9-YAP interaction.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Núcleo Celular , Fatores de Transcrição SOX9 , Fatores de Transcrição , Proteínas de Sinalização YAP , Humanos , Proteínas de Sinalização YAP/genética , Proteínas de Sinalização YAP/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Transporte Ativo do Núcleo Celular/genética , Camundongos , Linhagem Celular Tumoral , Animais , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...