Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.089
Filtrar
1.
Clin Transl Sci ; 17(7): e13881, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38982592

RESUMEN

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.


Asunto(s)
Apoptosis , Condrocitos , Clusterina , Interleucina-1beta , Osteoartritis de la Rodilla , Humanos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Osteoartritis de la Rodilla/patología , Osteoartritis de la Rodilla/metabolismo , Apoptosis/efectos de los fármacos , Clusterina/metabolismo , Clusterina/genética , Interleucina-1beta/metabolismo , Transducción de Señal/efectos de los fármacos , Células Cultivadas , Masculino , Persona de Mediana Edad , Anciano , Inflamación/metabolismo , Inflamación/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Femenino , Fosfatidilinositol 3-Quinasas/metabolismo , Morfolinas/farmacología , Cromonas/farmacología , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Mediadores de Inflamación/metabolismo , Óxido Nítrico/metabolismo
2.
BMC Musculoskelet Disord ; 25(1): 537, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997667

RESUMEN

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.


Asunto(s)
Exosomas , Interleucina-1beta , Degeneración del Disco Intervertebral , Núcleo Pulposo , Factor de Transcripción SOX9 , Humanos , Interleucina-1beta/metabolismo , Exosomas/metabolismo , Degeneración del Disco Intervertebral/patología , Degeneración del Disco Intervertebral/metabolismo , Degeneración del Disco Intervertebral/terapia , Núcleo Pulposo/metabolismo , Núcleo Pulposo/patología , Núcleo Pulposo/citología , Núcleo Pulposo/efectos de los fármacos , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Animales , Células Madre/metabolismo , Células Cultivadas , Agrecanos/metabolismo , Agrecanos/genética , Masculino , Orina/citología , Orina/química , Femenino , Colágeno Tipo II/metabolismo
3.
JCI Insight ; 9(11)2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38855864

RESUMEN

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.


Asunto(s)
Displasia Campomélica , Diferenciación Celular , Condrocitos , Condrogénesis , Factor de Transcripción SOX9 , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Animales , Condrocitos/metabolismo , Ratones , Displasia Campomélica/genética , Displasia Campomélica/patología , Displasia Campomélica/metabolismo , Condrogénesis/genética , Diferenciación Celular/genética , Elementos de Facilitación Genéticos/genética , Cromatina/metabolismo , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Ratones Noqueados , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/genética , Humanos , Desarrollo Óseo/genética
4.
Eur J Cell Biol ; 103(2): 151424, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38823166

RESUMEN

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.


Asunto(s)
Actinas , Desdiferenciación Celular , Condrocitos , Fibras de Estrés , Tropomiosina , Condrocitos/metabolismo , Condrocitos/citología , Fibras de Estrés/metabolismo , Animales , Actinas/metabolismo , Tropomiosina/metabolismo , Tropomiosina/genética , Fenotipo , Células Cultivadas , Proteína de Unión al GTP cdc42/metabolismo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Transactivadores/metabolismo , Transactivadores/genética
5.
Exp Cell Res ; 440(2): 114135, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38901791

RESUMEN

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.


Asunto(s)
Progresión de la Enfermedad , Células Estrelladas Hepáticas , Histonas , Cirrosis Hepática , Ratas Sprague-Dawley , Factor de Transcripción SOX9 , Animales , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Cirrosis Hepática/genética , Cirrosis Hepática/inducido químicamente , Histonas/metabolismo , Histonas/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Ratas , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Masculino , Proliferación Celular , Tetracloruro de Carbono , Factor de Crecimiento Transformador beta1/metabolismo , Humanos , Transcripción Genética , Movimiento Celular/genética , L-Lactato Deshidrogenasa/metabolismo , L-Lactato Deshidrogenasa/genética
6.
Osteoarthritis Cartilage ; 32(8): 872-885, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38852879

RESUMEN

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.


Asunto(s)
Estudio de Asociación del Genoma Completo , Factor 5 de Diferenciación de Crecimiento , Articulación de la Cadera , Osteoartritis de la Cadera , Polimorfismo de Nucleótido Simple , Humanos , Osteoartritis de la Cadera/genética , Osteoartritis de la Cadera/patología , Factor 5 de Diferenciación de Crecimiento/genética , Articulación de la Cadera/patología , Proteína Morfogenética Ósea 7/genética , Factor de Transcripción SOX9/genética , Predisposición Genética a la Enfermedad
7.
Sci Rep ; 14(1): 12297, 2024 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811798

RESUMEN

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.


Asunto(s)
Glándulas Suprarrenales , Factor de Transcripción SOX9 , Factor Esteroidogénico 1 , Animales , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Ratones , Factor Esteroidogénico 1/metabolismo , Factor Esteroidogénico 1/genética , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/embriología , Integrasas/metabolismo , Integrasas/genética , Ratones Noqueados , Femenino , Masculino
8.
Cancer Lett ; 593: 216939, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38729556

RESUMEN

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.


Asunto(s)
Aurora Quinasa A , Factor 4E Eucariótico de Iniciación , Infecciones por Helicobacter , Helicobacter pylori , Factor de Transcripción SOX9 , Neoplasias Gástricas , Animales , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Humanos , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Aurora Quinasa A/metabolismo , Aurora Quinasa A/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Factor 4E Eucariótico de Iniciación/genética , Ratones Noqueados , Ratones , Biosíntesis de Proteínas , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ubiquitinación
9.
Adv Sci (Weinh) ; 11(26): e2403227, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38704731

RESUMEN

To effectively treat osteoarthritis (OA), the existing inflammation must be reduced before the cartilage damage can be repaired; this cannot be achieved with a single type of extracellular vesicles (EVs). Here, a hydrogel complex with logic-gates function is proposed that can spatiotemporally controlled release two types of EVs: interleukin 10 (IL-10)+ EVs to promote M2 polarization of macrophage, and SRY-box transcription factor 9 (SOX9)+ EVs to increase cartilage matrix synthesis. Following dose-of-action screening, the dual EVs are loaded into a matrix metalloporoteinase 13 (MMP13)-sensitive self-assembled peptide hydrogel (KM13E) and polyethylene glycol diacrylate/gelatin methacryloyl-hydrogel microspheres (PGE), respectively. These materials are mixed to form a "microspheres-in-gel" KM13E@PGE system. In vitro, KM13E@PGE abruptly released IL-10+ EVs after 3 days and slowly released SOX9+ EVs for more than 30 days. In vivo, KM13E@PGE increased the CD206+ M2 macrophage proportion in the synovial tissue and decreased the tumor necrosis factor-α and IL-1ß levels. The aggrecan and SOX9 expressions in the cartilage tissues are significantly elevated following inflammation subsidence. This performance is not achieved using anti-inflammatory or cartilage repair therapy alone. The present study provides an injectable, integrated delivery system with spatiotemporal control release of dual EVs, and may inspire logic-gates strategies for OA treatment.


Asunto(s)
Modelos Animales de Enfermedad , Vesículas Extracelulares , Osteoartritis , Vesículas Extracelulares/metabolismo , Osteoartritis/metabolismo , Animales , Hidrogeles/química , Macrófagos/metabolismo , Interleucina-10/metabolismo , Humanos , Factor de Transcripción SOX9/metabolismo , Ratones , Ratas
10.
Differentiation ; 138: 100782, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38810379

RESUMEN

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.


Asunto(s)
Mandíbula , Cresta Neural , Animales , Embrión de Pollo , Ratones , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Tipificación del Cuerpo/genética , Cartílago/metabolismo , Cartílago/crecimiento & desarrollo , Cartílago/citología , Diferenciación Celular , Pollos/genética , Cilios/metabolismo , Cilios/genética , Regulación del Desarrollo de la Expresión Génica , Mandíbula/crecimiento & desarrollo , Mandíbula/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/crecimiento & desarrollo , Cresta Neural/citología , Cresta Neural/metabolismo , Transducción de Señal , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética
11.
J Oral Rehabil ; 51(8): 1507-1520, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38717032

RESUMEN

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.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II , Diferenciación Celular , Condrogénesis , Células Madre Mesenquimatosas , MicroARNs , Transducción de Señal , Estrés Mecánico , Membrana Sinovial , Humanos , Agrecanos/metabolismo , Agrecanos/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Diferenciación Celular/fisiología , Células Cultivadas , Condrogénesis/fisiología , Colágeno Tipo II/metabolismo , Colágeno Tipo II/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Membrana Sinovial/citología , Membrana Sinovial/metabolismo
12.
BMC Genomics ; 25(1): 464, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741085

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Cromatina , Gónadas , Factor de Transcripción SOX9 , Análisis de la Célula Individual , Animales , Cromatina/metabolismo , Cromatina/genética , Ratones , Linaje de la Célula/genética , Femenino , Masculino , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Gónadas/metabolismo , Gónadas/citología , Gónadas/embriología , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Proteínas WT1/genética , Proteínas WT1/metabolismo , Testículo/metabolismo , Testículo/citología , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Ovario/metabolismo , Ovario/citología
13.
PLoS One ; 19(5): e0301080, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38728328

RESUMEN

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.


Asunto(s)
Osteogénesis , Factor de Transcripción SOX9 , Articulación Temporomandibular , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Animales , Ratones , Articulación Temporomandibular/metabolismo , Articulación Temporomandibular/crecimiento & desarrollo , Osteogénesis/genética , Regulación hacia Abajo , Fibrocartílago/metabolismo , Ratones Transgénicos
14.
Int J Biol Sci ; 20(7): 2686-2697, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725852

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Células Madre Neoplásicas , Factor de Transcripción SOX9 , Ubiquitina-Proteína Ligasas , Femenino , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Lipopolisacáridos/farmacología , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Células Madre Neoplásicas/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba
15.
Int J Mol Sci ; 25(10)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38791601

RESUMEN

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.


Asunto(s)
Cartílago Articular , Proliferación Celular , Condrocitos , Osteoartritis , Osteofito , Humanos , Cartílago Articular/metabolismo , Cartílago Articular/patología , Cartílago Articular/cirugía , Condrocitos/metabolismo , Condrocitos/patología , Osteofito/metabolismo , Osteofito/patología , Masculino , Femenino , Anciano , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/cirugía , Persona de Mediana Edad , Colágeno Tipo II/metabolismo , Colágeno Tipo II/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Células Cultivadas , Movimiento Celular
16.
Nat Commun ; 15(1): 4148, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755149

RESUMEN

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.


Asunto(s)
Células Epiteliales Alveolares , Plasticidad de la Célula , Factor Nuclear Tiroideo 1 , Animales , Ratones , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/citología , Factor Nuclear Tiroideo 1/metabolismo , Factor Nuclear Tiroideo 1/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular , Epigénesis Genética , Ratones Endogámicos C57BL , Lesión Pulmonar/patología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/genética , Regeneración , Virus Sendai/genética , Virus Sendai/fisiología , Proliferación Celular , Ratones Noqueados , Pulmón/metabolismo
17.
Commun Biol ; 7(1): 545, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38714724

RESUMEN

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.


Asunto(s)
Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas , ARN Circular , Factor de Transcripción SOX9 , Neoplasias Gástricas , Factor de Transcripción 4 , beta Catenina , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Humanos , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , beta Catenina/metabolismo , beta Catenina/genética , ARN Circular/genética , ARN Circular/metabolismo , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Animales , Ratones , Línea Celular Tumoral , Ratones Desnudos , Masculino , Femenino , Resistencia a Antineoplásicos/genética , Ratones Endogámicos BALB C , Persona de Mediana Edad
18.
Sci Rep ; 14(1): 12508, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822021

RESUMEN

Adult vertebrate cartilage is usually quiescent. Some vertebrates possess ocular scleral skeletons composed of cartilage or bone. The morphological characteristics of the spotted wolffish (Anarhichas minor) scleral skeleton have not been described. Here we assessed the scleral skeletons of cultured spotted wolffish, a globally threatened marine species. The healthy spotted wolffish we assessed had scleral skeletons with a low percentage of cells staining for the chondrogenesis marker sex-determining region Y-box (Sox) 9, but harboured a population of intraocular cells that co-express immunoglobulin M (IgM) and Sox9. Scleral skeletons of spotted wolffish with grossly observable eye abnormalities displayed a high degree of perochondrial activation as evidenced by cellular morphology and expression of proliferating cell nuclear antigen (PCNA) and phosphotyrosine. Cells staining for cluster of differentiation (CD) 45 and IgM accumulated around sites of active chondrogenesis, which contained cells that strongly expressed Sox9. The level of scleral chondrogenesis and the numbers of scleral cartilage PCNA positive cells increased with the temperature of the water in which spotted wolffish were cultured. Our results provide new knowledge of differing Sox9 spatial tissue expression patterns during chondrogenesis in normal control and ocular insult paradigms. Our work also provides evidence that spotted wolffish possess an inherent scleral chondrogenesis response that may be sensitive to temperature. This work also advances the fundamental knowledge of teleost ocular skeletal systems.


Asunto(s)
Condrogénesis , Factor de Transcripción SOX9 , Animales , Factor de Transcripción SOX9/metabolismo , Esclerótica/metabolismo , Temperatura , Inmunoglobulina M/metabolismo , Ojo/metabolismo , Agua/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Cartílago/metabolismo
19.
Cell Stem Cell ; 31(5): 589-590, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701754

RESUMEN

Poorly regenerative organs deposit scar tissue to mend damage. Aggarwal et al. establish that transient Sox9 activity is necessary for early proximal tubule epithelial regeneration, while Trogisch et al. and Aggarwal et al. show that persistent Sox9 activity in epithelial and endothelial cells activates fibroblasts creating fibrotic microdomains in multiple organs.


Asunto(s)
Fibrosis , Factor de Transcripción SOX9 , Factor de Transcripción SOX9/metabolismo , Humanos , Animales , Fibroblastos/metabolismo , Fibroblastos/patología
20.
Arch Dermatol Res ; 316(5): 134, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662224

RESUMEN

Exploration of gene expression variations is a potential source to unravel biological pathways involved in pathological changes in body and understand the mechanism underneath. Vitiligo patients were explored for gene expression changes transcriptionally at perilesional site in comparison to normal site of same patients for melanogenesis pathway (TYR, DCT & TYRP1) cell adhesion (MMPs & TIMP1), cell survival (BCL2 & BAX1) as well as proliferation, migration & development (SOX9, SOX10 & MITF) regulatory system, using skin biopsy samples. Results were also compared with changes in gene expression for melanocytes under stress after hydrogen peroxide treatment in-vitro. Gene amplification was carried out via real time PCR. We found increased expression of proliferation, migration & development regulatory genes as well as melanogenesis pathway genes at perilesional site of patients. In-vitro study also supports induced MITF expression and disturbed melanogenesis in melanocytes under stress. Expression level ratio of cell survival regulatory genes' (BCL2/BAX1) as well as cell adhesion regulatory genes (MMPs/TIMP1) was observed upregulated at patient's perilesional site however downregulated in hydrogen peroxide treated melanocytes in-vitro. Observed upregulated gene expression at perilesional site of patients may be via positive feedback loop in response to stress to increase cell tolerance power to survive against adverse conditions. Gene expression analysis suggests better cell survival and proliferation potential at perilesional site in vitiligo patients. It seems in-vivo conditions/growth factors supports cells to fight for survival to accommodate stressed conditions.


Asunto(s)
Supervivencia Celular , Peróxido de Hidrógeno , Melanocitos , Vitíligo , Humanos , Vitíligo/genética , Vitíligo/patología , Melanocitos/metabolismo , Melanocitos/patología , Supervivencia Celular/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Masculino , Adulto , Femenino , Proliferación Celular/genética , Piel/patología , Piel/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Persona de Mediana Edad , Adulto Joven , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Biopsia , Adolescente , Adhesión Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA