Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21.320
1.
Arch Oral Biol ; 163: 105980, 2024 Jul.
Article En | MEDLINE | ID: mdl-38692246

OBJECTIVE: To determine the effect of hyaluronic acid (HA) degradation by hyaluronidase (HYAL) in inhibiting collagen fiber production by rat periodontal ligament cells (rPDLCs). DESIGN: Primary rPDLCs were isolated from the euthanized rats and used for in vitro experiments. The appropriate HYAL concentration was determined through CCK-8 testing for cytotoxicity detection and Alizarin red staining for mineralization detection. RT-qPCR and western blot assays were conducted to assess the effect of HYAL, with or without TGF-ß, on generation of collagen fiber constituents and expression of actin alpha 2, smooth muscle (ACTA2) of rPDLCs. RESULTS: Neither cell proliferation nor mineralization were significantly affected by treatment with 4 U/mL HYAL. HYAL (4 U/mL) alone downregulated type I collagen fiber (Col1a1 and Col1a2) and Acta2 mRNA expression; however, ACTA2 and COL1 protein levels were only downregulated by HYAL treatment after TGF-ß induction. CONCLUSIONS: Treatment of rPDLCs with HYAL can inhibit TGF-ß-induced collagen matrix formation and myofibroblast transformation.


Cell Proliferation , Collagen , Fibroblasts , Hyaluronoglucosaminidase , Myofibroblasts , Periodontal Ligament , Transforming Growth Factor beta , Animals , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Hyaluronoglucosaminidase/pharmacology , Rats , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Collagen/metabolism , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyaluronic Acid/pharmacology , Cells, Cultured , Rats, Sprague-Dawley , Actins/metabolism , Blotting, Western , In Vitro Techniques , Collagen Type I/metabolism , Biomarkers/metabolism , Real-Time Polymerase Chain Reaction , Male , RNA, Messenger/metabolism
2.
Med Oncol ; 41(6): 137, 2024 May 05.
Article En | MEDLINE | ID: mdl-38705933

Metastasis poses a significant challenge in combating tumors. Even in papillary thyroid cancer (PTC), which typically exhibits a favorable prognosis, high recurrence rates are attributed to metastasis. Cytoplasmic linker protein 170 (CLIP170) functions as a classical microtubule plus-end tracking protein (+TIP) and has shown close association with cell migration. Nevertheless, the specific impact of CLIP170 on PTC cells remains to be elucidated. Our analysis of the GEO and TCGA databases unveiled an association between CLIP170 and the progression of PTC. To explore the impact of CLIP170 on PTC cells, we conducted various assays. We evaluated its effects through CCK-8, wound healing assay, and transwell assay after knocking down CLIP170. Additionally, the influence of CLIP170 on the cellular actin structure was examined via immunofluorescence; we further investigated the molecular expressions of epithelial-mesenchymal transition (EMT) and the transforming growth factor-ß (TGF-ß) signaling pathways through Western blotting and RT-qPCR. These findings were substantiated through an in vivo nude mouse model of lung metastasis. We observed a decreased expression of CLIP170 in PTC in contrast to normal thyroid tissue. Functionally, the knockdown of CLIP170 (CLIP170KD) notably enhanced the metastatic potential and EMT of PTC cells, both in vitro and in vivo. Mechanistically, CLIP170KD triggered the activation of the TGF-ß pathway, subsequently promoting tumor cell migration, invasion, and EMT. Remarkably, the TGF-ß inhibitor LY2157299 effectively countered TGF-ß activity and significantly reversed tumor metastasis and EMT induced by CLIP170 knockdown. In summary, these findings collectively propose CLIP170 as a promising therapeutic target to mitigate metastatic tendencies in PTC.


Epithelial-Mesenchymal Transition , Microtubule-Associated Proteins , Neoplasm Proteins , Signal Transduction , Thyroid Cancer, Papillary , Thyroid Neoplasms , Transforming Growth Factor beta , Animals , Female , Humans , Male , Mice , Cell Line, Tumor , Cell Movement , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/genetics , Mice, Nude , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/genetics , Transforming Growth Factor beta/metabolism
3.
Commun Biol ; 7(1): 544, 2024 May 07.
Article En | MEDLINE | ID: mdl-38714800

Numerous myofibroblasts are arisen from endothelial cells (ECs) through endothelial to mesenchymal transition (EndMT) triggered by TGF-ß. However, the mechanism of ECs transforms to a different subtype, or whether there exists an intermediate state of ECs remains unclear. In present study, we demonstrate Midkine (MDK) mainly expressed by CD31 + ACTA2+ECs going through partial EndMT contribute greatly to myofibroblasts by spatial and single-cell transcriptomics. MDK is induced in TGF-ß treated ECs, which upregulates C/EBPß and increases EndMT genes, and these effects could be reversed by siMDK. Mechanistically, MDK promotes the binding ability of C/EBPß with ACTA2 promoter by stabilizing the C/EBPß protein. In vivo, knockout of Mdk or conditional knockout of Mdk in ECs reduces EndMT markers and significantly reverses fibrogenesis. In conclusion, our study provides a mechanistic link between the induction of EndMT by TGF-ß and MDK, which suggests that blocking MDK provides potential therapeutic strategies for renal fibrosis.


CCAAT-Enhancer-Binding Protein-beta , Fibrosis , Midkine , Midkine/metabolism , Midkine/genetics , Animals , Mice , Humans , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Epithelial-Mesenchymal Transition , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Transforming Growth Factor beta/metabolism , Mice, Inbred C57BL , Male , Kidney/metabolism , Kidney/pathology , Mice, Knockout , Endothelial-Mesenchymal Transition
4.
Genome Biol ; 25(1): 117, 2024 May 07.
Article En | MEDLINE | ID: mdl-38715110

BACKGROUND: Preeclampsia, one of the most lethal pregnancy-related diseases, is associated with the disruption of uterine spiral artery remodeling during placentation. However, the early molecular events leading to preeclampsia remain unknown. RESULTS: By analyzing placentas from preeclampsia, non-preeclampsia, and twin pregnancies with selective intrauterine growth restriction, we show that the pathogenesis of preeclampsia is attributed to immature trophoblast and maldeveloped endothelial cells. Delayed epigenetic reprogramming during early extraembryonic tissue development leads to generation of excessive immature trophoblast cells. We find reduction of de novo DNA methylation in these trophoblast cells results in selective overexpression of maternally imprinted genes, including the endoretrovirus-derived gene PEG10 (paternally expressed gene 10). PEG10 forms virus-like particles, which are transferred from the trophoblast to the closely proximate endothelial cells. In normal pregnancy, only a low amount of PEG10 is transferred to maternal cells; however, in preeclampsia, excessive PEG10 disrupts maternal vascular development by inhibiting TGF-beta signaling. CONCLUSIONS: Our study reveals the intricate epigenetic mechanisms that regulate trans-generational genetic conflict and ultimately ensure proper maternal-fetal interface formation.


Pre-Eclampsia , Trophoblasts , Vascular Remodeling , Pre-Eclampsia/genetics , Pregnancy , Female , Humans , Trophoblasts/metabolism , Vascular Remodeling/genetics , Placenta/metabolism , DNA Methylation , Epigenesis, Genetic , Endothelial Cells/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Imprinting , Transforming Growth Factor beta/metabolism , Fetal Growth Retardation/genetics , Placentation/genetics , RNA-Binding Proteins , Apoptosis Regulatory Proteins
5.
Elife ; 132024 May 28.
Article En | MEDLINE | ID: mdl-38805545

As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that CBFB (subunit of a heterodimeric Cbfß/Runx1, Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfb in tamoxifen-induced Cbfbf/f;Col2a1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfß deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/Yap signaling and Tgfß signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfß and Yap expression and increased active ß-catenin expression in articular cartilage, while local AAV-mediated Cbfb overexpression promoted Yap expression and diminished active ß-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfb overexpression in knee joints of mice with OA showed the significant protective effect of Cbfß on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfß may be the cause of OA. Moreover, Local admission of Cbfb may rescue and protect OA through decreasing Wnt/ß-catenin signaling, and increasing Hippo/Yap signaling and Tgfß/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfb overexpression could be an effective strategy for treatment of OA.


Cartilage, Articular , Hippo Signaling Pathway , Homeostasis , Osteoarthritis , Transforming Growth Factor beta , YAP-Signaling Proteins , Animals , Cartilage, Articular/metabolism , Mice , Osteoarthritis/genetics , Osteoarthritis/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Wnt Signaling Pathway , beta Catenin/metabolism , beta Catenin/genetics , Signal Transduction , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics
6.
Cell Death Dis ; 15(5): 368, 2024 May 28.
Article En | MEDLINE | ID: mdl-38806480

Transforming growth factor beta (TGFß) signaling plays a critical role in tumorigenesis and metastasis. However, little is known about the biological function of TGFbeta-induced lncRNA in cancer. In this study, we discovered a novel TGFbeta-induced lncRNA, termed TGILR, whose function in cancer remains unknown to date. TGILR expression was directly activated by the canonical TGFbeta/SMAD3 signaling axis, and this activation is highly conserved in cancer. Clinical analysis showed that TGILR overexpression showed a significant correlation with lymph node metastasis and poor survival and was an independent prognostic factor in gastric cancer (GC). Depletion of TGILR caused an obvious inhibitory effect on GC cell proliferation, invasion, and epithelial-mesenchymal transition (EMT) in vitro and in vivo. More importantly, we demonstrated that TGFbeta signaling in GC was overactivated due to cancer-associated fibroblast (CAF) infiltration. Mechanistically, increased level of CAF-secreted TGFbeta activates TGFbeta signaling, leading to TGILR overexpression in GC cells. Meanwhile, TGILR overexpression inhibited the microRNA biogenesis of miR-1306 and miR-33a by interacting with TARBP2 and reducing its protein stability, thereby promoting GC progression via TCF4-mediated EMT signaling. In conclusion, CAF infiltration drives GC metastasis and EMT signaling through activating TGFbeta/TGILR axis. Targeted blocking of CAF-derived TGFbeta should be a promising anticancer strategy in GC.


Cancer-Associated Fibroblasts , Disease Progression , Epithelial-Mesenchymal Transition , MicroRNAs , Signal Transduction , Stomach Neoplasms , Transforming Growth Factor beta , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Humans , Transforming Growth Factor beta/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Cell Proliferation , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Gene Expression Regulation, Neoplastic , Male , Mice, Nude , Female , Mice , Mice, Inbred BALB C , Smad3 Protein/metabolism
7.
Mol Biol Rep ; 51(1): 677, 2024 May 25.
Article En | MEDLINE | ID: mdl-38796641

BACKGROUND: One of the main causes of diabetic nephropathy is oxidative stress induced by hyperglycemia. Apelin inhibits insulin secretion. Besides, renal expression of TGF-ß is increased in diabetes mellitus (DM). The preventive effect of quercetin (Q) against renal functional disorders and tissue damage developed by DM in rats was assessed. METHODS: Forty male Wistar rats were grouped into normal control (NC), normal + quercetin (NQ: quercetin, 50 mg/kg/day by gavage), diabetic control (DC: streptozotocin, 65 mg/kg, i.p.), diabetic + quercetin pretreatment (D + Qpre), and diabetic + quercetin post-treatment (D + Qpost). All samples (24-hour urine, plasma, pancreatic, and renal tissues) were obtained at the terminal of the experiment. RESULTS: Compared to NC and NQ groups, DM ended in elevated plasma and glucose levels, decreased plasma insulin level, kidney dysfunction, augmented levels of malondialdehyde, decreased level of reduced glutathione, reduced enzymatic activities of superoxide dismutase and catalase, elevated gene expression levels of apelin and TGF-ß, also renal and pancreatic histological damages. Quercetin administration diminished entire the changes. However, the measure of improvement in the D + Qpre group was higher than that of the D + Qpost group. CONCLUSION: Quercetin prevents renal dysfunction induced by DM, which might be related to the diminution of lipid peroxidation, strengthening of antioxidant systems, and prevention of the apelin/ TGF-ß signaling pathway.


Apelin , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Kidney , Oxidative Stress , Quercetin , Rats, Wistar , Transforming Growth Factor beta , Animals , Quercetin/pharmacology , Rats , Male , Transforming Growth Factor beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/prevention & control , Diabetic Nephropathies/drug therapy , Apelin/metabolism , Oxidative Stress/drug effects , Blood Glucose/metabolism , Blood Glucose/drug effects , Antioxidants/pharmacology , Antioxidants/metabolism , Insulin/metabolism , Insulin/blood , Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation/drug effects
8.
Cell Commun Signal ; 22(1): 296, 2024 May 28.
Article En | MEDLINE | ID: mdl-38807115

BACKGROUND: The SARS-CoV-2 virus causes severe COVID-19 in one-fifth of patients. In addition to high mortality, infection may induce respiratory failure and cardiovascular complications associated with inflammation. Acute or prolonged inflammation results in organ fibrosis, the cause of which might be endothelial disorders arising during the endothelial-mesenchymal transition (EndMT). METHODS: HUVECs and HMEC-1 cells were stimulated with SARS-CoV-2 S (Spike) and N (Nucleocapsid) proteins, and EndMT induction was evaluated by studying specific protein markers via Western blotting. Wound healing and tube formation assays were employed to assess the potential of SARS-CoV-2 to stimulate changes in cell behaviour. MRTF nuclear translocation, ROS generation, TLR4 inhibitors, TGF-ß-neutralizing antibodies, and inhibitors of the TGF-ß-dependent pathway were used to investigate the role of the TGF-ß-MRTF signalling axis in SARS-CoV-2-dependent EndMT stimulation. RESULTS: Both viral proteins stimulate myofibroblast trans-differentiation. However, the N protein is more effective at EndMT induction. The TGF-ß-MRTF pathway plays a critical role in this process. The N protein preferentially favours action through TGF-ß2, whose secretion is induced through TLR4-ROS action. TGF-ß2 stimulates MRTF-A and MRTF-B nuclear translocation and strongly regulates EndMT. In contrast, the Spike protein stimulates TGF-ß1 secretion as a result of ACE2 downregulation. TGF-ß1 induces only MRTF-B, which, in turn, weakly regulates EndMT. Furthermore, aspirin, a common nonsteroidal anti-inflammatory drug, might prevent and reverse SARS-CoV-2-dependent EndMT induction through TGF-ß-MRTF pathway deregulation. CONCLUSION: The reported study revealed that SARS-CoV-2 infection induces EndMT. Moreover, it was demonstrated for the first time at the molecular level that the intensity of the EndMT triggered by SARS-CoV-2 infection may vary and depend on the viral protein involved. The N protein acts through TLR4-ROS-TGF-ß2-MRTF-A/B, whereas the S protein acts through ACE2-TGF-ß1-MRTF-B. Furthermore, we identified aspirin as a potential anti-fibrotic drug for treating patients with SARS-CoV-2 infection.


Aspirin , COVID-19 , Coronavirus Nucleocapsid Proteins , Epithelial-Mesenchymal Transition , SARS-CoV-2 , Signal Transduction , Spike Glycoprotein, Coronavirus , Transforming Growth Factor beta , Humans , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Transforming Growth Factor beta/metabolism , COVID-19/metabolism , COVID-19/virology , Coronavirus Nucleocapsid Proteins/metabolism , Aspirin/pharmacology , Signal Transduction/drug effects , Epithelial-Mesenchymal Transition/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Transcription Factors/metabolism , Toll-Like Receptor 4/metabolism , Cell Line , Endothelial-Mesenchymal Transition , Phosphoproteins
9.
Sci Rep ; 14(1): 11553, 2024 05 21.
Article En | MEDLINE | ID: mdl-38773312

Knee osteoarthritis is a chronic joint disease mainly characterized by cartilage degeneration. The treatment is challenging due to the lack of blood vessels and nerve supplies in cartilaginous tissue, causing a prominent limitation of regenerative capacity. Hence, we investigated the cellular promotional and anti-inflammatory effects of sericin, Bombyx mori-derived protein, on three-dimensional chondrogenic ATDC5 cell models. The results revealed that a high concentration of sericin promoted chondrogenic proliferation and differentiation and enhanced matrix production through the increment of glycosaminoglycans, COL2A1, COL X, and ALP expressions. SOX-9 and COL2A1 gene expressions were notably elevated in sericin treatment. The proteomic analysis demonstrated the upregulation of phosphoglycerate mutase 1 and triosephosphate isomerase, a glycolytic enzyme member, reflecting the proliferative enhancement of sericin. The differentiation capacity of sericin was indicated by the increased expressions of procollagen12a1, collagen10a1, rab1A, periostin, galectin-1, and collagen6a3 proteins. Sericin influenced the differentiation capacity via the TGF-ß signaling pathway by upregulating Smad2 and Smad3 while downregulating Smad1, BMP2, and BMP4. Importantly, sericin exhibited an anti-inflammatory effect by reducing IL-1ß, TNF-α, and MMP-1 expressions and accelerating COL2A1 production in the early inflammatory stage. In conclusion, sericin demonstrates potential in promoting chondrogenic proliferation and differentiation, enhancing cartilaginous matrix synthesis through glycolysis and TGF-ß signaling pathways, and exhibiting anti-inflammatory properties.


Cell Differentiation , Cell Proliferation , Chondrogenesis , Glycolysis , Inflammation , Sericins , Signal Transduction , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Smad2 Protein/metabolism , Animals , Signal Transduction/drug effects , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Chondrogenesis/drug effects , Sericins/pharmacology , Glycolysis/drug effects , Mice , Inflammation/metabolism , Inflammation/pathology , Inflammation/drug therapy , Chondrocytes/metabolism , Chondrocytes/drug effects , Cell Line , Bombyx/metabolism
10.
Cancer Lett ; 592: 216953, 2024 Jun 28.
Article En | MEDLINE | ID: mdl-38729557

TGFBR2, a key regulator of the TGFß signaling pathway, plays a crucial role in gastric cancer (GC) metastasis through its endosomal recycling process. Despite its importance, the mechanisms governing this process remain unclear. Here, we identify integrin ß5 (ITGB5) as a critical mediator that promotes TGFBR2 endosomal recycling. Our study reveals elevated expression of ITGB5 in GC, particularly in metastatic cases, correlating with poor patient outcomes. Knockdown of ITGB5 impairs GC cell metastasis both in vitro and in vivo. Mechanistically, ITGB5 facilitates epithelial-mesenchymal transition mediated by TGFß signaling, thereby enhancing GC metastasis. Acting as a scaffold, ITGB5 interacts with TGFBR2 and SNX17, facilitating SNX17-mediated endosomal recycling of TGFBR2 and preventing lysosomal degradation, thereby maintaining its surface distribution on tumor cells. Notably, TGFß signaling directly upregulates ITGB5 expression, establishing a positive feedback loop that exacerbates GC metastasis. Our findings shed light on the role of ITGB5 in promoting GC metastasis through SNX17-mediated endosomal recycling of TGFBR2, providing insights for the development of targeted cancer therapies.


Endosomes , Epithelial-Mesenchymal Transition , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Stomach Neoplasms , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Humans , Endosomes/metabolism , Animals , Cell Line, Tumor , Integrin beta Chains/metabolism , Integrin beta Chains/genetics , Sorting Nexins/genetics , Sorting Nexins/metabolism , Mice , Neoplasm Metastasis , Gene Expression Regulation, Neoplastic , Transforming Growth Factor beta/metabolism
11.
J Cell Mol Med ; 28(10): e18400, 2024 May.
Article En | MEDLINE | ID: mdl-38780513

Osteosarcoma is the most common primary bone malignancy in children and adolescents. Overexpression of polo-like kinase 1 (PLK1) is frequent in osteosarcoma and drives disease progression and metastasis, making it a promising therapeutic target. In this study, we explored PLK1 knockdown in osteosarcoma cells using RNA interference mediated by high-fidelity Cas13d (hfCas13d). PLK1 was found to be significantly upregulated in osteosarcoma tumour tissues compared to normal bone. sgRNA-mediated PLK1 suppression via hfCas13d transfection inhibited osteosarcoma cell proliferation, induced G2/M cell cycle arrest, promoted apoptosis, reduced cell invasion and increased expression of the epithelial marker E-cadherin. Proximity labelling by TurboID coupled with co-immunoprecipitation identified novel PLK1 interactions with Smad3, a key intracellular transducer of TGF-ß signalling. PLK1 knockdown impaired Smad2/3 phosphorylation and modulated TGF-ß/Smad3 pathway inactivation. Finally, in vivo delivery of hfCas13d vectors targeting PLK1 substantially attenuated osteosarcoma xenograft growth in nude mice. Taken together, this study highlights PLK1 as a potential therapeutic target and driver of disease progression in osteosarcoma. It also demonstrates the utility of hfCas13d-mediated gene knockdown as a strategy for targeted therapy. Further optimization of PLK1 suppression approaches may ultimately improve clinical outcomes for osteosarcoma patients.


Apoptosis , Cell Cycle Proteins , Cell Proliferation , Mice, Nude , Osteosarcoma , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , RNA Interference , Signal Transduction , Smad3 Protein , Transforming Growth Factor beta , Osteosarcoma/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Animals , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Transforming Growth Factor beta/metabolism , Mice , Apoptosis/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , Xenograft Model Antitumor Assays , Female
12.
Cell Commun Signal ; 22(1): 257, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711089

Benign prostatic hyperplasia (BPH) is a multifactorial disease in which abnormal growth factor activation and embryonic reawakening are considered important factors. Here we demonstrated that the aberrant activation of transforming growth factor ß (TGF-ß)/Rho kinase 1 (ROCK1) increased the stemness of BPH tissue by recruiting mesenchymal stem cells (MSCs), indicating the important role of embryonic reawakening in BPH. When TGF-ß/ROCK1 is abnormally activated, MSCs are recruited and differentiate into fibroblasts/myofibroblasts, leading to prostate stromal hyperplasia. Further research showed that inhibition of ROCK1 activation suppressed MSC migration and their potential for stromal differentiation. Collectively, our findings suggest that abnormal activation of TGF-ß/ROCK1 regulates stem cell lineage specificity, and the small molecule inhibitor GSK269962A could target ROCK1 and may be a potential treatment for BPH.


Mesenchymal Stem Cells , Prostatic Hyperplasia , Transforming Growth Factor beta , rho-Associated Kinases , rho-Associated Kinases/metabolism , Male , Prostatic Hyperplasia/pathology , Prostatic Hyperplasia/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Prostate/pathology , Prostate/metabolism , Cell Movement , Mice , Stromal Cells/metabolism , Stromal Cells/pathology
13.
Sci Adv ; 10(18): eadj8042, 2024 May 03.
Article En | MEDLINE | ID: mdl-38691608

Overactivation of the transforming growth factor-ß (TGFß) signaling in Duchenne muscular dystrophy (DMD) is a major hallmark of disease progression, leading to fibrosis and muscle dysfunction. Here, we investigated the role of SETDB1 (SET domain, bifurcated 1), a histone lysine methyltransferase involved in muscle differentiation. Our data show that, following TGFß induction, SETDB1 accumulates in the nuclei of healthy myotubes while being already present in the nuclei of DMD myotubes where TGFß signaling is constitutively activated. Transcriptomics revealed that depletion of SETDB1 in DMD myotubes leads to down-regulation of TGFß target genes coding for secreted factors involved in extracellular matrix remodeling and inflammation. Consequently, SETDB1 silencing in DMD myotubes abrogates the deleterious effect of their secretome on myoblast differentiation by impairing myoblast pro-fibrotic response. Our findings indicate that SETDB1 potentiates the TGFß-driven fibrotic response in DMD muscles, providing an additional axis for therapeutic intervention.


Histone-Lysine N-Methyltransferase , Muscle Fibers, Skeletal , Muscular Dystrophy, Duchenne , Signal Transduction , Transforming Growth Factor beta , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Transforming Growth Factor beta/metabolism , Humans , Animals , Cell Differentiation , Mice , Myoblasts/metabolism , Fibrosis , Gene Expression Regulation
14.
Sci Rep ; 14(1): 10910, 2024 05 13.
Article En | MEDLINE | ID: mdl-38740884

Transforming growth factor-ß (TGF-ß) signaling plays a significant role in multiple biological processes, including inflammation, immunity, and cell death. However, its specific impact on the cochlea remains unclear. In this study, we aimed to investigate the effects of TGF-ß signaling suppression on auditory function and cochlear pathology in mice with kanamycin-induced ototoxicity. Kanamycin and furosemide (KM-FS) were systemically administered to 8-week-old C57/BL6 mice, followed by immediate topical application of a TGF-ß receptor inhibitor (TGF-ßRI) onto the round window membrane. Results showed significant TGF-ß receptor upregulation in spiral ganglion neurons (SGNs) after KM-FA ototoxicity, whereas expression levels in the TGF-ßRI treated group remained unchanged. Interestingly, despite no significant change in cochlear TGF-ß expression after KM-FS ototoxicity, TGF-ßRI treatment resulted in a significant decrease in TGF-ß signaling. Regarding auditory function, TGF-ßRI treatment offered no therapeutic effects on hearing thresholds and hair cell survival following KM-FS ototoxicity. However, SGN loss and macrophage infiltration were significantly increased with TGF-ßRI treatment. These results imply that inhibition of TGF-ß signaling after KM-FS ototoxicity promotes cochlear inflammation and SGN degeneration.


Kanamycin , Mice, Inbred C57BL , Ototoxicity , Signal Transduction , Spiral Ganglion , Transforming Growth Factor beta , Animals , Kanamycin/toxicity , Signal Transduction/drug effects , Ototoxicity/etiology , Ototoxicity/metabolism , Ototoxicity/pathology , Transforming Growth Factor beta/metabolism , Mice , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Spiral Ganglion/pathology , Cochlea/metabolism , Cochlea/drug effects , Cochlea/pathology , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Furosemide/pharmacology , Male
15.
Support Care Cancer ; 32(6): 365, 2024 May 17.
Article En | MEDLINE | ID: mdl-38758235

BACKGROUND: Cancer-related depression is a well-documented condition that significantly impacts long-term quality of life. Brain-derived neurotrophic factor (BDNF), a neurotrophin essential for neurogenesis and neuronal plasticity, has been implicated in various neuropsychological disorders including depression associated with cancer. Cytokines, on the other hand, play a crucial role in regulating depression, potentially by influencing BDNF expression. Transforming growth factor-ß (TGF-ß), a key immune regulator within the tumor microenvironment, has been found to elevate BDNF levels, establishing a link between peripheral immune responses and depression. The study aims to investigate the correlation of TGF-ß and BDNF in cancer-related depression. METHODS: This study involved a cohort of 153 gynecological patients, including 61 patients with gynecological cancer and 92 patients without cancer. Depression levels were assessed using the subscale of Hospital Anxiety and Depression Scale (HADS-D), and TGF-ß and BDNF plasma levels were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS: The study revealed elevated plasma TGF-ß levels in patients with cancer (32.24 ± 22.93 ng/ml) compared to those without cancer (25.24 ± 19.72 ng/ml) (P = 0.046). Additionally, reduced levels of BDNF were observed in patients presenting depression symptoms (44.96 ± 41.06 pg/ml) compared to those without depression (133.5 ± 176.7 pg/ml) (P = 0.036). Importantly, a significant correlation between TGF-ß and BDNF was found in patients without cancer but with depression (correlation coefficient = 0.893, **P < 0.01). Interestingly, cancer appeared to influence the association between TGF-ß and BDNF in patients with depression, as evidenced by a significant difference in the correlation of TGF-ß and BDNF between cancer and non-cancer groups (P = 0.041). CONCLUSIONS: These findings underscore the active involvement of TGF-ß and BDNF crosstalk in the context of cancer-related depression.


Brain-Derived Neurotrophic Factor , Depression , Transforming Growth Factor beta , Humans , Brain-Derived Neurotrophic Factor/blood , Female , Cross-Sectional Studies , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism , Depression/etiology , Middle Aged , Adult , Genital Neoplasms, Female/complications , Genital Neoplasms, Female/psychology , Quality of Life , Enzyme-Linked Immunosorbent Assay , Aged , Psychiatric Status Rating Scales , Case-Control Studies
16.
BMC Gastroenterol ; 24(1): 163, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745150

BACKGROUND: The liver regeneration is a highly complicated process depending on the close cooperations between the hepatocytes and non-parenchymal cells involving various inflammatory cells. Here, we explored the role of myeloid-derived suppressor cells (MDSCs) in the processes of liver regeneration and liver fibrosis after liver injury. METHODS: We established four liver injury models of mice including CCl4-induced liver injury model, bile duct ligation (BDL) model, concanavalin A (Con A)-induced hepatitis model, and lipopolysaccharide (LPS)-induced hepatitis model. The intrahepatic levels of MDSCs (CD11b+Gr-1+) after the liver injury were detected by flow cytometry. The effects of MDSCs on liver tissues were analyzed in the transwell co-culture system, in which the MDSCs cytokines including IL-10, VEGF, and TGF-ß were measured by ELISA assay and followed by being blocked with specific antibodies. RESULTS: The intrahepatic infiltrations of MDSCs with surface marker of CD11b+Gr-1+ remarkably increased after the establishment of four liver injury models. The blood served as the primary reservoir for hepatic recruitment of MDSCs during the liver injury, while the bone marrow appeared play a compensated role in increasing the number of MDSCs at the late stage of the inflammation. The recruited MDSCs in injured liver were mainly the M-MDSCs (CD11b+Ly6G-Ly6Chigh) featured by high expression levels of cytokines including IL-10, VEGF, and TGF-ß. Co-culture of the liver tissues with MDSCs significantly promoted the proliferation of both hepatocytes and hepatic stellate cells (HSCs). CONCLUSIONS: The dramatically and quickly infiltrated CD11b+Gr-1+ MDSCs in injured liver not only exerted pro-proliferative effects on hepatocytes, but also accounted for the activation of profibrotic HSCs.


CD11b Antigen , Liver Cirrhosis , Liver Regeneration , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells , Animals , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/immunology , Mice , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Regeneration/physiology , CD11b Antigen/metabolism , Male , Disease Models, Animal , Liver/pathology , Liver/metabolism , Vascular Endothelial Growth Factor A/metabolism , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Concanavalin A , Ligation , Lipopolysaccharides , Interleukin-10/metabolism , Transforming Growth Factor beta/metabolism , Hepatic Stellate Cells/metabolism , Coculture Techniques , Hepatocytes/metabolism , Hepatocytes/pathology , Bile Ducts
17.
J Clin Invest ; 134(10)2024 Mar 26.
Article En | MEDLINE | ID: mdl-38747285

Transforming growth factor ß (TGF-ß) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-ß remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-ß in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-ß. The activation of latent TGF-ß requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-ß, rebalanced TGF-ß signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-ß in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.


Fibroblasts , Fibrosis , Transforming Growth Factor beta , Wnt-5a Protein , rho-Associated Kinases , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Animals , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Mice , Humans , Fibroblasts/metabolism , Fibroblasts/pathology , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Scleroderma, Systemic/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/genetics , Mice, Knockout , Wnt Proteins/metabolism , Wnt Proteins/genetics , MAP Kinase Signaling System , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/genetics
18.
PLoS One ; 19(5): e0302662, 2024.
Article En | MEDLINE | ID: mdl-38748716

Kaab Dum, a prominent indigenous rice variety cultivated in the Pak Phanang Basin of Nakhon Si Thammarat, Thailand, is the focus of our study. We investigate the therapeutic potential of indigenous Kaab Dum rice extract in the context of chronic wounds. Our research encompasses an examination of the nutritional compositions and chemical profiles of Kaab Dum rice extract. Additionally, we assess how the extract affects chronic wounds in TGF-ß-induced HaCaT cells. Our evaluation methods include the detection of cellular oxidative stress, the examination of endoplasmic reticulum (ER) stress, wound healing assays, analysis of cell cycle arrest and the study of cellular senescence through senescence-associated ß-galactosidase (SA-ß-gal) staining. Our research findings demonstrate that TGF-ß induces oxidative stress in HaCaT cells, which subsequently triggers ER stress, confirmed by the expression of the PERK protein. This ER stress results in cell cycle arrest in HaCaT cells, characterized by an increase in p21 protein, a cyclin-dependent kinase inhibitor (CDKI). Ultimately, this leads to cellular senescence, as confirmed by SA-ß-gal staining. Importantly, our study reveals the effectiveness of Kaab Dum rice extract in promoting wound healing in the chronic wound model. The extract reduces ER stress and senescent cells. These beneficial effects are potentially linked to the antioxidant and anti-inflammatory properties of the rice extract. The findings of our study have the potential to make significant contributions to the development of enhanced products for both the prevention and treatment of chronic wounds.


Cellular Senescence , Endoplasmic Reticulum Stress , Keratinocytes , Oryza , Plant Extracts , Wound Healing , Humans , Oryza/chemistry , Cellular Senescence/drug effects , Wound Healing/drug effects , Endoplasmic Reticulum Stress/drug effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Plant Extracts/pharmacology , Thailand , Cell Line , HaCaT Cells , Oxidative Stress/drug effects , Transforming Growth Factor beta/metabolism , Cell Cycle Checkpoints/drug effects , Southeast Asian People
19.
Pak J Pharm Sci ; 37(1): 33-41, 2024 Jan.
Article En | MEDLINE | ID: mdl-38741398

The objective of this research is to assess how salvianolate impacts inflammation and oxidative stress in a laboratory setting, as well as to investigate the underlying mechanisms. HK-2 cells were subjected to different treatments, including normal glucose, mannitol, high glucose and high glucose plus salvianolate. Cell proliferation, death, MDA levels, IL-1ß, IL-6, TNF-α, MCP-1 concentrations, ROS levels, MMP, MPTP and ATP levels were assessed using various kits. The protein expressions of NOX4, TGF-ß1, P-Smad2, P-Smad3, Smad4 and Smad7 were ascertained through western blot analysis. Our results indicated salvianolate could reduce the release of IL-1ß, IL-6, TNF-α, as well as MCP-1, alleviate the levels of oxidative stress markers NOX4 and MDA, and improve mitochondrial function by increasing MMP and ATP levels while reducing ROS and MPTP opening. Furthermore, salvianolate inhibited the TGF-ß1/Smad2, Smad3 signaling pathway, suppressed Smad4 expression and increased Smad7 expression. Salvianolate seems to mitigate inflammation and oxidative stress through a variety of mechanisms. These discoveries offer valuable understanding into the possible mechanisms by which salvianolate may be employed in the treatment of diabetic nephropathy.


Glucose , Inflammation , Oxidative Stress , Signal Transduction , Oxidative Stress/drug effects , Signal Transduction/drug effects , Glucose/metabolism , Humans , Cell Line , Inflammation/drug therapy , Inflammation/metabolism , Smad Proteins/metabolism , Plant Extracts/pharmacology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/pharmacology , Cell Proliferation/drug effects
20.
Invest Ophthalmol Vis Sci ; 65(5): 15, 2024 May 01.
Article En | MEDLINE | ID: mdl-38717426

Purpose: Mutations in the genes encoding type IV collagen alpha 1 (COL4A1) and alpha 2 (COL4A2) cause a multisystem disorder that includes ocular anterior segment dysgenesis (ASD) and glaucoma. We previously showed that transforming growth factor beta (TGFß) signaling was elevated in developing anterior segments from Col4a1 mutant mice and that reducing TGFß signaling ameliorated ASD, supporting a role for the TGFß pathway in disease pathogenesis. Here, we tested whether altered TGFß signaling also contributes to glaucoma-related phenotypes in Col4a1 mutant mice. Methods: To test the role of TGFß signaling in glaucoma-relevant phenotypes, we genetically reduced TGFß signaling using mice with mutated Tgfbr2, which encodes the common receptor for all TGFß ligands in Col4a1+/G1344D mice. We performed slit-lamp biomicroscopy and optical coherence tomography for qualitative and quantitative analyses of anterior and posterior ocular segments, histological analyses of ocular tissues and optic nerves, and intraocular pressure assessments using rebound tonometry. Results: Col4a1+/G1344D mice showed defects of the ocular drainage structures, including iridocorneal adhesions, and phenotypes consistent with glaucomatous neurodegeneration, including thinning of the nerve fiber layer, retinal ganglion cell loss, optic nerve head excavation, and optic nerve degeneration. We found that reducing TGFß receptor 2 (TGFBR2) was protective for ASD, ameliorated ocular drainage structure defects, and protected against glaucomatous neurodegeneration in Col4a1+/G1344D mice. Conclusions: Our results suggest that elevated TGFß signaling contributes to glaucomatous neurodegeneration in Col4a1 mutant mice.


Collagen Type IV , Glaucoma , Intraocular Pressure , Receptor, Transforming Growth Factor-beta Type II , Signal Transduction , Tomography, Optical Coherence , Transforming Growth Factor beta , Animals , Mice , Collagen Type IV/metabolism , Collagen Type IV/genetics , Signal Transduction/physiology , Intraocular Pressure/physiology , Glaucoma/metabolism , Glaucoma/genetics , Glaucoma/pathology , Transforming Growth Factor beta/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Disease Models, Animal , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/genetics , Mice, Inbred C57BL , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Anterior Eye Segment/metabolism , Anterior Eye Segment/pathology , Optic Nerve/pathology , Optic Nerve/metabolism , Slit Lamp Microscopy , Phenotype , Tonometry, Ocular , Mutation
...