Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.684
Filter
1.
BMC Cancer ; 24(1): 883, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039505

ABSTRACT

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.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Movement , Cell Proliferation , Exosomes , Lung Neoplasms , Mesenchymal Stem Cells , MicroRNAs , SOX9 Transcription Factor , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , Exosomes/metabolism , Exosomes/genetics , Mice , Cell Proliferation/genetics , Mesenchymal Stem Cells/metabolism , Cell Movement/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Male , Female , A549 Cells , Xenograft Model Antitumor Assays
2.
BMC Vet Res ; 20(1): 326, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39026327

ABSTRACT

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.


Subject(s)
Animal Feed , Cichlids , Dietary Supplements , Methyltestosterone , Mucuna , Tribulus , Animals , Male , Tribulus/chemistry , Methyltestosterone/pharmacology , Animal Feed/analysis , Mucuna/chemistry , Cichlids/growth & development , Cichlids/genetics , Plant Extracts/pharmacology , Plant Extracts/chemistry , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Diet/veterinary , Forkhead Box Protein L2/genetics , Female , Testosterone/blood
3.
BMC Musculoskelet Disord ; 25(1): 537, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997667

ABSTRACT

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.


Subject(s)
Exosomes , Interleukin-1beta , Intervertebral Disc Degeneration , Nucleus Pulposus , SOX9 Transcription Factor , Humans , Interleukin-1beta/metabolism , Exosomes/metabolism , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/therapy , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Nucleus Pulposus/cytology , Nucleus Pulposus/drug effects , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Animals , Stem Cells/metabolism , Cells, Cultured , Aggrecans/metabolism , Aggrecans/genetics , Male , Urine/cytology , Urine/chemistry , Female , Collagen Type II/metabolism
4.
Clin Transl Sci ; 17(7): e13881, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38982592

ABSTRACT

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.


Subject(s)
Apoptosis , Chondrocytes , Clusterin , Interleukin-1beta , Osteoarthritis, Knee , Humans , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/metabolism , Apoptosis/drug effects , Clusterin/metabolism , Clusterin/genetics , Interleukin-1beta/metabolism , Signal Transduction/drug effects , Cells, Cultured , Male , Middle Aged , Aged , Inflammation/metabolism , Inflammation/pathology , Proto-Oncogene Proteins c-akt/metabolism , Female , Phosphatidylinositol 3-Kinases/metabolism , Morpholines/pharmacology , Chromones/pharmacology , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Matrix Metalloproteinase 13/metabolism , Inflammation Mediators/metabolism , Nitric Oxide/metabolism
5.
Mol Biol Rep ; 51(1): 862, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073659

ABSTRACT

BACKGROUND: Understanding how healthy articular cartilage responds to mechanical loading is critical. Moderate mechanical loading has positive effects on the cartilage, such as maintaining cartilage homeostasis. The degree of mechanical loading is determined by a combination of intensity, frequency, and duration; however, the best combination of these parameters for knee cartilage remains unclear. This study aimed to determine which combination of intensity, frequency, and duration provides the best mechanical loading on healthy knee articular cartilage in vitro and in vivo. METHODS AND RESULTS: In this study, 33 male mice were used. Chondrocytes isolated from mouse knee joints were subjected to different cyclic tensile strains (CTSs) and assessed by measuring the expression of cartilage matrix-related genes. Furthermore, the histological characteristics of mouse tibial cartilages were quantified using different treadmill exercises. Chondrocytes and mice were divided into the control group and eight intervention groups: high-intensity, high-frequency, and long-duration; high-intensity, high-frequency, and short-duration; high-intensity, low-frequency, and long-duration; high-intensity, low-frequency, and short-duration; low-intensity, high-frequency, and long-duration; low-intensity, high-frequency, and short-duration; low-intensity, low-frequency, and long-duration; low-intensity, low-frequency, and short-duration. In low-intensity CTSs, chondrocytes showed anabolic responses by altering the mRNA expression of COL2A1 in short durations and SOX9 in long durations. Furthermore, low-intensity, low-frequency, and long-duration treadmill exercises minimized chondrocyte hypertrophy and enhanced aggrecan synthesis in tibial cartilages. CONCLUSION: Low-intensity, low-frequency, and long-duration mechanical loading is the best combination for healthy knee cartilage to maintain homeostasis and activate anabolic responses. Our findings provide a significant scientific basis for exercise and lifestyle instructions.


Subject(s)
Cartilage, Articular , Chondrocytes , Stress, Mechanical , Weight-Bearing , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/physiology , Mice , Chondrocytes/metabolism , Male , Weight-Bearing/physiology , Physical Conditioning, Animal/physiology , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Collagen Type II/metabolism , Collagen Type II/genetics , Knee Joint/metabolism , Knee Joint/physiology , Mice, Inbred C57BL
6.
JCI Insight ; 9(11)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38855864

ABSTRACT

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.


Subject(s)
Campomelic Dysplasia , Cell Differentiation , Chondrocytes , Chondrogenesis , SOX9 Transcription Factor , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Animals , Chondrocytes/metabolism , Mice , Campomelic Dysplasia/genetics , Campomelic Dysplasia/pathology , Campomelic Dysplasia/metabolism , Chondrogenesis/genetics , Cell Differentiation/genetics , Enhancer Elements, Genetic/genetics , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation, Developmental , Mice, Knockout , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Humans , Bone Development/genetics
7.
Eur J Cell Biol ; 103(2): 151424, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38823166

ABSTRACT

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.


Subject(s)
Actins , Cell Dedifferentiation , Chondrocytes , Stress Fibers , Tropomyosin , Chondrocytes/metabolism , Chondrocytes/cytology , Stress Fibers/metabolism , Animals , Actins/metabolism , Tropomyosin/metabolism , Tropomyosin/genetics , Phenotype , Cells, Cultured , cdc42 GTP-Binding Protein/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Trans-Activators/metabolism , Trans-Activators/genetics
8.
Exp Cell Res ; 440(2): 114135, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38901791

ABSTRACT

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.


Subject(s)
Disease Progression , Hepatic Stellate Cells , Histones , Liver Cirrhosis , Rats, Sprague-Dawley , SOX9 Transcription Factor , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/chemically induced , Histones/metabolism , Histones/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Rats , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Male , Cell Proliferation , Carbon Tetrachloride , Transforming Growth Factor beta1/metabolism , Humans , Transcription, Genetic , Cell Movement/genetics , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics
9.
J Clin Invest ; 134(15)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38885336

ABSTRACT

Osteogenesis imperfecta (OI) type V is the second most common form of OI, distinguished by hyperplastic callus formation and calcification of the interosseous membranes, in addition to the bone fragility. It is caused by a recurrent, dominant pathogenic variant (c.-14C>T) in interferon-induced transmembrane protein 5 (IFITM5). Here, we generated a conditional Rosa26-knockin mouse model to study the mechanistic consequences of the recurrent mutation. Expression of the mutant Ifitm5 in osteo-chondroprogenitor or chondrogenic cells resulted in low bone mass and growth retardation. Mutant limbs showed impaired endochondral ossification, cartilage overgrowth, and abnormal growth plate architecture. The cartilage phenotype correlates with the pathology reported in patients with OI type V. Surprisingly, expression of mutant Ifitm5 in mature osteoblasts caused no obvious skeletal abnormalities. In contrast, earlier expression in osteo-chondroprogenitors was associated with an increase in the skeletal progenitor cell population within the periosteum. Lineage tracing showed that chondrogenic cells expressing the mutant Ifitm5 had decreased differentiation into osteoblastic cells in diaphyseal bone. Moreover, mutant IFITM5 disrupted early skeletal homeostasis in part by activating ERK signaling and downstream SOX9 protein, and inhibition of these pathways partially rescued the phenotype in mutant animals. These data identify the contribution of a signaling defect altering osteo-chondroprogenitor differentiation as a driver in the pathogenesis of OI type V.


Subject(s)
Cell Differentiation , MAP Kinase Signaling System , Osteoblasts , Osteogenesis Imperfecta , SOX9 Transcription Factor , Animals , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Mice , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Osteogenesis Imperfecta/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Humans , Mutation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Osteogenesis/genetics , Mice, Transgenic , Stem Cells/metabolism , Stem Cells/pathology , Mitogen-Activated Protein Kinase 3
10.
Colloids Surf B Biointerfaces ; 241: 114011, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38838445

ABSTRACT

Hyaline cartilage regeneration will bring evangel to millions of people suffered from cartilage diseases. However, uncontrollable cartilage fibrosis and matrix mineralization are the primary causes of cartilage regeneration failure in many tissue engineering scaffolds. This study presents a new attempt to avoid endochondral ossification or fibrosis in cartilage regeneration therapy by establishing biochemical regulatory area. Here, SOX9 expression plasmids are assembled in cellulose gels by chitosan gene vectors to fabricate SOX9+ functionalized scaffolds. RT-qPCR, western blot and biochemical analysis all show that the SOX9 reinforcement strategy can enhance chondrogenic specific proteins expression and promote GAG production. Notably, the interference from SOX9 has resisted osteogenic inducing significantly, showing an inhibition of COL1, OPN and OC production, and the inhibition efficiency was about 58.4 %, 22.8 % and 76.9 % respectively. In vivo study, implantation of these scaffolds with BMSCs can induce chondrogenic differentiation and resist endochondral ossification effectively. Moreover, specific SOX9+ functionalized area of the gel exhibited the resistance to matrix mineralization, indicating the special biochemical functional area for cartilage regeneration. These results indicate that this strategy is effective for promoting the hyaline cartilage regeneration and avoiding cartilage fibrosis, which provides a new insight to the future development of cartilage regeneration scaffolds.


Subject(s)
Chondrogenesis , Fibrosis , SOX9 Transcription Factor , Tissue Scaffolds , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Tissue Scaffolds/chemistry , Animals , Cartilage/metabolism , Cartilage/pathology , Tissue Engineering , Cell Differentiation , Chitosan/chemistry , Humans , Cells, Cultured , Osteogenesis/drug effects , Regeneration
11.
Cell Stem Cell ; 31(5): 589-590, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38701754

ABSTRACT

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.


Subject(s)
Fibrosis , SOX9 Transcription Factor , SOX9 Transcription Factor/metabolism , Humans , Animals , Fibroblasts/metabolism , Fibroblasts/pathology
12.
Cancer Lett ; 593: 216939, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38729556

ABSTRACT

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.


Subject(s)
Aurora Kinase A , Eukaryotic Initiation Factor-4E , Helicobacter Infections , Helicobacter pylori , SOX9 Transcription Factor , Stomach Neoplasms , Animals , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/genetics , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Humans , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Aurora Kinase A/metabolism , Aurora Kinase A/genetics , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4E/genetics , Mice, Knockout , Mice , Protein Biosynthesis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Ubiquitination
13.
PLoS One ; 19(5): e0301080, 2024.
Article in English | MEDLINE | ID: mdl-38728328

ABSTRACT

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.


Subject(s)
Osteogenesis , SOX9 Transcription Factor , Temporomandibular Joint , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Animals , Mice , Temporomandibular Joint/metabolism , Temporomandibular Joint/growth & development , Osteogenesis/genetics , Down-Regulation , Fibrocartilage/metabolism , Mice, Transgenic
14.
Sci Rep ; 14(1): 12297, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811798

ABSTRACT

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.


Subject(s)
Adrenal Glands , SOX9 Transcription Factor , Steroidogenic Factor 1 , Animals , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Mice , Steroidogenic Factor 1/metabolism , Steroidogenic Factor 1/genetics , Adrenal Glands/metabolism , Adrenal Glands/embryology , Integrases/metabolism , Integrases/genetics , Mice, Knockout , Female , Male
15.
Int J Biol Sci ; 20(7): 2686-2697, 2024.
Article in English | MEDLINE | ID: mdl-38725852

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Neoplastic Stem Cells , SOX9 Transcription Factor , Ubiquitin-Protein Ligases , Female , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Lipopolysaccharides/pharmacology , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Neoplastic Stem Cells/metabolism , NF-kappa B/metabolism , Signal Transduction , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Up-Regulation
16.
J Oral Rehabil ; 51(8): 1507-1520, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38717032

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II , Cell Differentiation , Chondrogenesis , Mesenchymal Stem Cells , MicroRNAs , Signal Transduction , Stress, Mechanical , Synovial Membrane , Humans , Aggrecans/metabolism , Aggrecans/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Cell Differentiation/physiology , Cells, Cultured , Chondrogenesis/physiology , Collagen Type II/metabolism , Collagen Type II/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Signal Transduction/physiology , Smad Proteins/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Synovial Membrane/cytology , Synovial Membrane/metabolism
17.
Differentiation ; 138: 100782, 2024.
Article in English | MEDLINE | ID: mdl-38810379

ABSTRACT

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.


Subject(s)
Mandible , Neural Crest , Animals , Chick Embryo , Mice , Avian Proteins/genetics , Avian Proteins/metabolism , Body Patterning/genetics , Cartilage/metabolism , Cartilage/growth & development , Cartilage/cytology , Cell Differentiation , Chickens/genetics , Cilia/metabolism , Cilia/genetics , Gene Expression Regulation, Developmental , Mandible/growth & development , Mandible/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mesoderm/growth & development , Neural Crest/cytology , Neural Crest/metabolism , Signal Transduction , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics
18.
Adv Sci (Weinh) ; 11(26): e2403227, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704731

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Extracellular Vesicles , Osteoarthritis , Extracellular Vesicles/metabolism , Osteoarthritis/metabolism , Animals , Hydrogels/chemistry , Macrophages/metabolism , Interleukin-10/metabolism , Humans , SOX9 Transcription Factor/metabolism , Mice , Rats
19.
In Vitro Cell Dev Biol Anim ; 60(6): 609-615, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38727898

ABSTRACT

Osteoblast-derived semaphorin3A (Sema3A) has been reported to be involved in bone protection, and Sema3A knockout mice have been reported to exhibit chondrodysplasia. From these reports, Sema3A is considered to be involved in chondrogenic differentiation and skeletal formation, but there are many unclear points about its function and mechanism in chondrogenic differentiation. This study investigated the pharmacological effects of Sema3A in chondrogenic differentiation. The amount of Sema3A secreted into the culture supernatant was measured using an enzyme-linked immunosorbent assay. The expression of chondrogenic differentiation-related factors, such as Type II collagen (COL2A1), Aggrecan (ACAN), hyaluronan synthase 2 (HAS2), SRY-box transcription factor 9 (Sox9), Runt-related transcription factor 2 (Runx2), and Type X collagen (COL10A1) in ATDC5 cells treated with Sema3A (1,10 and 100 ng/mL) was examined using real-time reverse transcription polymerase chain reaction. Further, to assess the deposition of total glycosaminoglycans during chondrogenic differentiation, ATDC5 cells were stained with Alcian Blue. Moreover, the amount of hyaluronan in the culture supernatant was measured by enzyme-linked immunosorbent assay. The addition of Sema3A to cultured ATDC5 cells increased the expression of Sox9, Runx2, COL2A1, ACAN, HAS2, and COL10A1 during chondrogenic differentiation. Moreover, it enhanced total proteoglycan and hyaluronan synthesis. Further, Sema3A was upregulated in the early stages of chondrogenic differentiation, and its secretion decreased later. Sema3A increases extracellular matrix production and promotes chondrogenic differentiation. To the best of our knowledge, this is the first study to demonstrate the role of Sema3A on chondrogenic differentiation.


Subject(s)
Cell Differentiation , Chondrogenesis , Semaphorin-3A , Animals , Cell Differentiation/drug effects , Semaphorin-3A/metabolism , Chondrogenesis/drug effects , Mice , Chondrocytes/metabolism , Chondrocytes/cytology , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Cell Line , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Collagen Type II/metabolism , Collagen Type II/genetics , Aggrecans/metabolism , Aggrecans/genetics , Hyaluronan Synthases/metabolism , Hyaluronan Synthases/genetics , Glycosaminoglycans/metabolism , Collagen Type X/metabolism , Collagen Type X/genetics
20.
BMC Genomics ; 25(1): 464, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741085

ABSTRACT

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.


Subject(s)
Cell Lineage , Chromatin , Gonads , SOX9 Transcription Factor , Single-Cell Analysis , Animals , Chromatin/metabolism , Chromatin/genetics , Mice , Cell Lineage/genetics , Female , Male , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Gonads/metabolism , Gonads/cytology , Gonads/embryology , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , Testis/metabolism , Testis/cytology , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Ovary/metabolism , Ovary/cytology
SELECTION OF CITATIONS
SEARCH DETAIL