Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.574
Filter
1.
Mol Biol Rep ; 51(1): 1031, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352573

ABSTRACT

BACKGROUND: Fatty liver disease is a metabolic disorder that recently has been classified into two categories: metabolic dysfunction-associated fatty liver disease (MAFLD) and non-MAFLD. TGF-ß signaling pathway is likely a significant factor in the pathogenesis of this condition, exerting its effects through its downstream signaling proteins, Smad2/3. Accordingly, this study aimed to investigate the TGF-ß signaling pathway in the white blood cells (WBCs) of patients with MAFLD compared to those with non-MAFLD and control groups. METHODS AND RESULTS: In this study, 41 patients with fatty liver were evaluated, comprising 22 patients with MAFLD and 19 patients with non-MAFLD, and compared to 22 healthy controls. Gene expression of TGF-ß1, TGF-ß3, and CTGF were quantified using qRT-PCR, and the protein expressions of Smad2/3 and P-Smad2/3 were analyzed using western blotting. Gene expression analysis revealed a significant decrease in the gene expressions of the TGF-ß1 and TGF-ß3 and an increase in CTGF gene expression in patients with MAFLD and non-MAFLD compared to the control group. Notably, the Smad2/3 protein expression was significantly higher in the non-MAFLD group compared to the control group (P < 0.05). On the other hand, the P-smad2/3 protein expression was significantly elevated in the MAFLD group compared to the control group (P < 0.001). CONCLUSIONS: TGF-ß signaling pathway in WBCs of patients with fatty liver are affected by a complex signaling pathway. However, metabolic factors most probably affect TGF-ß1 gene expression and its downstream signaling proteins more than TGF-ß3.


Subject(s)
Connective Tissue Growth Factor , Signal Transduction , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta1 , Humans , Male , Case-Control Studies , Female , Middle Aged , Adult , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Fatty Liver/metabolism , Fatty Liver/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Smad Proteins/metabolism , Smad Proteins/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Gene Expression Regulation
2.
Int J Mol Sci ; 25(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39273564

ABSTRACT

Cellular Communication Network Factor 2 (CCN2) is a matricellular protein implicated in cell communication and microenvironmental signaling. Overexpression of CCN2 has been documented in various cardiovascular pathologies, wherein it may exert either deleterious or protective effects depending on the pathological context, thereby suggesting that its role in the cardiovascular system is not yet fully elucidated. In this study, we aimed to investigate the effects of Ccn2 gene deletion on the progression of acute cardiac injury induced by doxorubicin (DOX), a widely utilized chemotherapeutic agent. To this end, we employed conditional knockout (KO) mice for the Ccn2 gene (CCN2-KO), which were administered DOX and compared to DOX-treated wild-type (WT) control mice. Our findings demonstrated that the ablation of CCN2 ameliorated DOX-induced cardiac dysfunction, as evidenced by improvements in ejection fraction (EF) and fractional shortening (FS) of the left ventricle. Furthermore, DOX-treated CCN2-KO mice exhibited a significant reduction in the gene expression and activation of oxidative stress markers (Hmox1 and Nfe2l2/NRF2) relative to DOX-treated WT controls. Additionally, the deletion of Ccn2 markedly attenuated DOX-induced cardiac fibrosis. Collectively, these results suggest that CCN2 plays a pivotal role in the pathogenesis of DOX-mediated cardiotoxicity by modulating oxidative stress and fibrotic pathways. These findings provide a novel avenue for future investigations to explore the therapeutic potential of targeting CCN2 in the prevention of DOX-induced cardiac dysfunction.


Subject(s)
Connective Tissue Growth Factor , Doxorubicin , Fibrosis , Mice, Knockout , Oxidative Stress , Animals , Doxorubicin/adverse effects , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Mice , Oxidative Stress/drug effects , Gene Deletion , Male , Myocardium/metabolism , Myocardium/pathology , Mice, Inbred C57BL , Cardiotoxicity/genetics , Cardiotoxicity/metabolism
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167490, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39236363

ABSTRACT

Vascular endothelial inflammation is crucial in hepatic ischemia-reperfusion injury (IRI). Our previous research has shown that connective tissue growth factor (CTGF), secreted by endothelial cells, protects against acute liver injury, but its upstream mechanism is unclear. We aimed to clarify the protective role of CTGF in endothelial cell inflammation during IRI and reveal the regulation between endoplasmic reticulum stress-induced activating transcription factor 6 (ATF6) and CTGF. Hypoxia/reoxygenation in endothelial cells, hepatic IRI in mice and clinical specimens were used to examine the relationships between CTGF and inflammatory factors and determine how ATF6 regulates CTGF and reduces damage. We found that activating ATF6 promoted CTGF expression and reduced liver damage in hepatic IRI. In vitro, activated ATF6 upregulated CTGF and downregulated inflammation, while ATF6 inhibition had the opposite effect. Dual-luciferase assays and chromatin immunoprecipitation confirmed that activated ATF6 binds to the CTGF promoter, enhancing its expression. Activated ATF6 increases CTGF and reduces extracellular regulated protein kinase 1/2 (ERK1/2) phosphorylation, decreasing inflammatory factors. Conversely, inhibiting ATF6 decreases CTGF and increases the phosphorylation of ERK1/2, increasing inflammatory factor levels. ERK1/2 inhibition reverses this effect. Clinical samples have shown that CTGF increases after IRI, inversely correlating with inflammatory cytokines. Therefore, ATF6 activation during liver IRI enhances CTGF expression and reduces endothelial inflammation via ERK1/2 inhibition, providing a novel target for diagnosing and treating liver IRI.


Subject(s)
Activating Transcription Factor 6 , Connective Tissue Growth Factor , Liver , Reperfusion Injury , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Animals , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Humans , Mice , Male , Liver/metabolism , Liver/pathology , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Endoplasmic Reticulum Stress/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , MAP Kinase Signaling System/drug effects
4.
PLoS One ; 19(9): e0309833, 2024.
Article in English | MEDLINE | ID: mdl-39240867

ABSTRACT

Pulmonary fibrosis (PF) is an interstitial lung disease characterized by inflammation and fibrotic changes, with an unknown cause. In the early stages of PF, severe inflammation leads to the destruction of lung tissue, followed by upregulation of fibrotic factors like Transforming growth factor-ß (TGF-ß) and connective tissue growth factor (CTGF), which disrupt normal tissue repair. Geniposide, a natural iridoid glycoside primarily derived from the fruits of Gardenia jasminoides Ellis, possesses various pharmacological activities, including liver protection, choleretic effects, and anti-inflammatory properties. In this study, we investigated the effects of Geniposide on chronic inflammation and fibrosis induced by bleomycin (BLM) in mice with pulmonary fibrosis (PF). PF was induced by intratracheal instillation of bleomycin, and Geniposide(100/50/25mg•kg-1) was orally administered to the mice once a day until euthanasia(14 day/28 day). The Raw264.7 cell inflammation induced by LPS was used to evaluate the effect of Geniposide on the activation of macrophage. Our results demonstrated that Geniposide reduced lung coefficients, decreased the content of Hydroxyproline, and improved pathological changes in lung tissue. It also reduced the number of inflammatory cells and levels of pro-inflammatory cytokines in bronchoalveolar lavage fluid (BALF) of bleomycin-induced PF mice. At the molecular level, Geniposide significantly down-regulated the expression of TGF-ß1, Smad2/3, p38, and CTGF in lung tissues of PF mice induced by bleomycin. Molecular docking results revealed that Geniposide exhibited good binding activity with TGF-ß1, Smad2, Smad3, and p38. In vitro study showed Geniposide directly inhibited the activation of macrophage induced by LPS. In conclusion, our findings suggest that Geniposide can ameliorate bleomycin-induced pulmonary fibrosis in mice by inhibiting the TGF-ß/Smad and p38MAPK signaling pathways.


Subject(s)
Bleomycin , Iridoids , Pulmonary Fibrosis , Transforming Growth Factor beta , p38 Mitogen-Activated Protein Kinases , Animals , Bleomycin/adverse effects , Bleomycin/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Iridoids/pharmacology , Mice , p38 Mitogen-Activated Protein Kinases/metabolism , Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Male , RAW 264.7 Cells , Lung/pathology , Lung/drug effects , Lung/metabolism , Smad Proteins/metabolism , Connective Tissue Growth Factor/metabolism , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL
5.
Mol Biol Rep ; 51(1): 950, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39222158

ABSTRACT

BACKGROUND: Hepatic fibrosis, a prevalent chronic liver condition, involves excessive extracellular matrix production associated with aberrant wound healing. Hepatic stellate cells (HSCs) play a pivotal role in liver fibrosis, activated by inflammatory factors such as sphingosine 1-phosphate (S1P). Despite S1P's involvement in fibrosis, its specific role and downstream pathway in HSCs remain controversial. METHODS: In this study, we investigated the regulatory role of S1P/S1P receptor (S1PR) in Hippo-YAP activation in both LX-2 cell lines and primary HSCs. Real-time PCR, western blot, pharmacological inhibitors, siRNAs, and Rho activity assays were adopted to address the molecular mechanisms of S1P mediated YAP activation. RESULTS: Serum and exogenous S1P significantly increased the expression of YAP target genes in HSCs. Pharmacologic inhibitors and siRNA-mediated knockdowns of S1P receptors showed S1P receptor 2 (S1PR2) as the primary mediator for S1P-induced CTGF expression in HSCs. Results using siRNA-mediated knockdown, Verteporfin, and Phospho-Tag immunoblots showed that S1P-S1PR2 signaling effectively suppressed the Hippo kinases cascade, thereby activating YAP. Furthermore, S1P increased RhoA activities in cells and ROCK inhibitors effectively blocked CTGF induction. Cytoskeletal-perturbing reagents were shown to greatly modulate CTGF induction, suggesting the important role of actin cytoskeleton in S1P-induced YAP activation. Exogeneous S1P treatment was enough to increase the expression of COL1A1 and α-SMA, that were blocked by YAP specific inhibitor. CONCLUSIONS: Our data demonstrate that S1P/S1PR2-Src-RhoA-ROCK axis leads to Hippo-YAP activation, resulting in the up-regulation of CTGF, COL1A1 and α-SMA expression in HSCs. Therefore, S1PR2 may represent a potential therapeutic target for hepatic fibrosis.


Subject(s)
Connective Tissue Growth Factor , Hepatic Stellate Cells , Lysophospholipids , Signal Transduction , Sphingosine , Transcription Factors , YAP-Signaling Proteins , rho-Associated Kinases , rhoA GTP-Binding Protein , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Lysophospholipids/metabolism , Lysophospholipids/pharmacology , Humans , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Sphingosine/analogs & derivatives , Sphingosine/metabolism , YAP-Signaling Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Cell Line , Liver Cirrhosis/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Receptors, Lysosphingolipid/metabolism , Receptors, Lysosphingolipid/genetics , Collagen Type I/metabolism , Collagen Type I/genetics , Hippo Signaling Pathway
6.
Zhongguo Zhong Yao Za Zhi ; 49(17): 4702-4710, 2024 Sep.
Article in Chinese | MEDLINE | ID: mdl-39307818

ABSTRACT

This study aims to investigate the effects of Linggui Zhugan Decoction(LGZGD) on myocardial fibrosis(MF) and the Lats1/Yap signaling pathway in mice after myocardial infarction(MI), exploring its role and mechanism in inhibiting MF. The MI-induced ischemic mouse model was established by left anterior descending coronary artery ligation, followed by continuous intervention for six weeks. Doppler ultrasound imaging-system of small animals was used to detect left ventricular ejection fraction(LVEF), left ventricular fractional shortening(LVFS), left ventricular internal diameter at end-systole(LVIDs), and left ventricular internal diameter at end-diastole(LVIDd). Pathological changes in myocardial tissue were observed by HE and Masson staining. Serum levels of creatine kinase isoenzyme MB(CK-MB) and lactate dehydrogenase(LDH) were detected by using ELISA. Myocardial tissue mRNA levels of Lats1, Yap, and connective tissue growth factor(CTGF) were determined by RT-qPCR. Protein expression of alpha-smooth muscle actin(α-SMA), collagen Ⅰ(Col Ⅰ), collagen Ⅲ(Col Ⅲ), tissue inhibitor of metal protease 1(TIMP1), matrix metallopeptidase 2(MMP2), Yap, p-Yap, and n-Yap was determined by Western blot. Compared with the sham group, the model group showed significantly decreased LVEF and LVFS levels, increased LVIDd and LVIDs levels(P<0.01), disordered arrangement of myocardial cells, partial fracture of myocardial fibers, and massive deposition of collagen fibers. Moreover, serum levels of CK-MB and LDH were significantly increased(P<0.01), while myocardial tissue mRNA levels of Lats1 were significantly decreased(P<0.01), and mRNA levels of Yap and CTGF were significantly increased(P<0.01). Protein expression of α-SMA, Col Ⅰ, Col Ⅲ, MMP2, Yap, and n-Yap was significantly increased(P<0.01), while protein expression of Lats1, TIMP1, p-Yap, and the ratio of p-Yap/Yap were significantly decreased(P<0.01). Compared with the model group, after intervention with LGZGD(9.36 g·kg~(-1)), mice showed significantly increased LVEF and LVFS levels, decreased LVIDd and LVIDs levels(P<0.01), more orderly arrangement of myocardial cells, significantly reduced myocardial fiber fracture and collagen fiber deposition. Serum levels of CK-MB and LDH were significantly decreased(P<0.01), while myocardial tissue mRNA levels of Lats1 were significantly increased(P<0.01), and mRNA levels of Yap and CTGF were significantly decreased(P<0.01). Protein expression of α-SMA, Col Ⅰ, Col Ⅲ, MMP2, Yap, and n-Yap was significantly decreased(P<0.01), while protein expression of Lats1, TIMP1, p-Yap, and the ratio of p-Yap/Yap were significantly increased(P<0.01). LGZGD can inhibit MF in mice after MI and improve mouse cardiac function, which is closely related to the activation of the Lats1/Yap signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing , Drugs, Chinese Herbal , Fibrosis , Myocardial Infarction , Myocardium , Protein Serine-Threonine Kinases , Signal Transduction , YAP-Signaling Proteins , Animals , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Mice, Inbred C57BL , Humans , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics
7.
Clin Exp Pharmacol Physiol ; 51(9): e13913, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103233

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by chronic inflammation, lung tissue fibrotic changes and impaired lung function. Pulmonary fibrosis 's pathological process is thought to be influenced by macrophage-associated phenotypes. IPF treatment requires specific targets that target macrophage polarization. Cytokine-like 1(CYTL1) is a secreted protein with multiple biological functions first discovered in CD34+ haematopoietic cells. However, its possible effects on IPF progression remain unclear. This study investigated the role of CYTL1 in IPF progression in a bleomycin-induced lung injury and fibrosis model. In bleomycin-induced mice, CYTL1 is highly expressed. Moreover, CYTL1 ablation alleviates lung injury and fibrosis in vivo. Further, downregulating CYTL1 reduces macrophage M2 polarization. Mechanically, CYTL1 regulates transforming growth factor ß (TGF-ß)/connective tissue growth factor (CCN2) axis and inhibition of TGF-ß pathway alleviates bleomycin-induced lung injury and fibrosis. In conclusion, highly expressed CYTL1 inhibits macrophage M2 polarization by regulating TGF-ß/CCN2 expression, alleviating bleomycin-induced lung injury and fibrosis. CYTL1 could, therefore, serve as a promising IPF target.


Subject(s)
Bleomycin , Connective Tissue Growth Factor , Down-Regulation , Macrophages , Pulmonary Fibrosis , Transforming Growth Factor beta , Animals , Bleomycin/toxicity , Mice , Down-Regulation/drug effects , Transforming Growth Factor beta/metabolism , Macrophages/metabolism , Macrophages/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Mice, Inbred C57BL , Male , Cell Polarity/drug effects , Signal Transduction/drug effects , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology
8.
J Cell Mol Med ; 28(16): e70025, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39164826

ABSTRACT

Metastasis is a crucial stage in tumour progression, and cancer-associated fibroblasts (CAFs) support metastasis through their participation in extracellular matrix (ECM) stiffness. CD248 is a possible biomarker for non-small cell lung cancer (NSCLC)-derived CAFs, but its role in mediating ECM stiffness to promote NSCLC metastasis is unknown. We investigated the significance of CD248+ CAFs in activating the Hippo axis and promoting connective tissue growth factor (CTGF) expression, which affects the stromal collagen I environment and improves ECM stiffness, thereby facilitating NSCLC metastasis. In this study, we found that higher levels of CD248 in CAFs induced the formation of collagen I, which in turn increased extracellular matrix stiffness, thereby enabling NSCLC cell infiltration and migration. Hippo axis activation by CD248+ CAFs induces CTGF expression, which facilitates the formation of the collagen I milieu in the stromal matrix. In a tumour lung metastasis model utilizing fibroblast-specific CD248 gene knockout mice, CD248 gene knockout mice showed a significantly reduced ability to develop tumour lung metastasis compared to that of WT mice. Our findings demonstrate that CD248+ CAFs activate the Hippo pathway, thereby inducing CTGF expression, which in turn facilitates the collagen I milieu of the stromal matrix, which promotes NSCLC metastasis.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Non-Small-Cell Lung , Connective Tissue Growth Factor , Extracellular Matrix , Hippo Signaling Pathway , Lung Neoplasms , Mice, Knockout , Protein Serine-Threonine Kinases , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Animals , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Extracellular Matrix/metabolism , Mice , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cell Line, Tumor , Antigens, CD/metabolism , Antigens, CD/genetics , Neoplasm Metastasis , Signal Transduction , Gene Expression Regulation, Neoplastic , Cell Movement , Tumor Microenvironment
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(4): 886-892, 2024 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-39170023

ABSTRACT

Objective: To investigate the effect of the loss of myeloid-derived growth factor (Mydgf) on the transformation of cardiac fibroblasts into myofibroblasts after myocardial infarction (MI). Methods: Two adult mouse groups, including a wild-type (WT) group and another group with Mydgf knockout (Mydgf-KO), were examined in the study. The mice in these two groups were tested for their cardiac function by measuring left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) (n=10). Quantitative real-time PCR (qRT-PCR) (n=3) was performed to determine the mRNA expression levels of myofibroblast markers, including α-smooth muscle actin (α-SMA), periostin (postn), type Ⅷ collagen (col8al), and connective tissue growth factor (ctgf). Western blot (n=3) was performed to verify the protein expression levels of α-SMA. MI modeling was performed on the WT and the Mydgf-KO mice. Postoperative LVEF and LVFS (n=10) were then measured. The hearts were harvested and Masson staining was performed to determine the infarcted area (n=10). The heart samples of Mydgf-KO and WT mice were collected at d 7 and d 14 after MI, respectively, to verify the expression of myofibroblast markers (n=3). Results: Compared with WT mice, LVEF and LVFS in adult Mydgf-KO mice showed no significant changes (all P>0.05). However, the mRNA levels of α-SMA and postn were upregulated, and α-SMA protein expression was also increased (all P<0.05). After MI, compared with WT mice, LVEF and LVFS in Mydgf-KO mice decreased, and the infarcted area increased significantly (all P<0.05). Furthermore, mRNA levels of α-SMA, col8al, postn, and ctgf were increased in Mydgf-KO mice. In addition, the α-SMA protein expression level was upregulated and α-SMA-positive fibroblasts were increased (P<0.05). Conclusion: Mydgf deletion promotes the transformation of cardiac fibroblasts into myofibroblasts and aggravates myocardial fibrosis after MI.


Subject(s)
Connective Tissue Growth Factor , Fibrosis , Mice, Knockout , Myocardial Infarction , Myofibroblasts , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Mice , Myofibroblasts/metabolism , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Actins/metabolism , Actins/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Myocardium/metabolism , Myocardium/pathology , Mice, Inbred C57BL , Male , Fibroblasts/metabolism
10.
J Agric Food Chem ; 72(35): 19413-19423, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39178398

ABSTRACT

Chicken is the main source of protein for humans in most parts of the world. However, excessive fat deposition in chickens has become a serious problem. This adversely affects the growth of chickens and causes economic losses. Fat formation mainly occurs through preadipocyte differentiation, and excessive fat deposition results from the accumulation of preadipocytes after differentiation. Our previous studies have found that the connective tissue growth factor (CTGF) may be an important candidate gene for fat deposition. However, its function and mechanism in preadipocyte differentiation are still unclear. In this study, the RT-qPCR and Western blot results showed that the expression of CTGF mRNA and protein in the abdominal adipose of lean chickens was significantly higher than that of fat chickens. Therefore, we studied the function and mechanism of the CTGF in the differentiation of chicken preadipocytes. Functionally, the CTGF inhibited the differentiation of chicken preadipocytes. Mechanistically, the CTGF mediated the TGFß1/Smad3 signaling pathway, thereby inhibiting the differentiation of chicken preadipocytes. In addition, we used the unique molecular identifier (UMI) RNA-Seq technology to detect genes that can be regulated by the CTGF in the whole genome. Through transcriptome data analysis, we selected actin gamma 2 (ACTG2) as a candidate gene. Regarding the function of the ACTG2 gene, we found that it inhibited the differentiation of chicken preadipocytes. Furthermore, we found that the CTGF can inhibit the differentiation of preadipocytes through the ACTG2 gene. In summary, this study found the CTGF as a new negative regulator of chicken preadipocyte differentiation. The results of this study help improve the understanding of the molecular genetic mechanism of chicken adipose tissue growth and development and also have reference significance for the study of human obesity.


Subject(s)
Adipocytes , Cell Differentiation , Chickens , Connective Tissue Growth Factor , Signal Transduction , Smad3 Protein , Animals , Chickens/genetics , Chickens/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Adipocytes/metabolism , Adipocytes/cytology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Adipogenesis , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics
11.
Sci Rep ; 14(1): 18752, 2024 08 13.
Article in English | MEDLINE | ID: mdl-39138242

ABSTRACT

Subretinal fibrosis is a major untreatable cause of poor outcomes in neovascular age-related macular degeneration. Mouse models of subretinal fibrosis all possess a degree of invasiveness and tissue damage not typical of fibrosis progression. This project characterises JR5558 mice as a model to study subretinal fibrosis. Fundus and optical coherence tomography (OCT) imaging was used to non-invasively track lesions. Lesion number and area were quantified with ImageJ. Retinal sections, wholemounts and Western blots were used to characterise alterations. Subretinal lesions expand between 4 and 8 weeks and become established in size and location around 12 weeks. Subretinal lesions were confirmed to be fibrotic, including various cell populations involved in fibrosis development. Müller cell processes extended from superficial retina into subretinal lesions at 8 weeks. Western blotting revealed increases in fibronectin (4 wk and 8 wk, p < 0.001), CTGF (20 wks, p < 0.001), MMP2 (12 wks and 20 wks p < 0.05), αSMA (12 wks and 20 wks p < 0.05) and GFAP (8 wk and 12 wk, p ≤ 0.01), consistent with our immunofluorescence results. Intravitreal injection of Aflibercept reduced subretinal lesion growth. Our study provides evidence JR5558 mice have subretinal fibrotic lesions that grow between 4 and 8 weeks and confirms this line to be a good model to study subretinal fibrosis development and assess treatment options.


Subject(s)
Disease Models, Animal , Fibrosis , Retina , Tomography, Optical Coherence , Animals , Mice , Tomography, Optical Coherence/methods , Retina/pathology , Retina/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Fibronectins/metabolism , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Macular Degeneration/pathology , Macular Degeneration/metabolism , Matrix Metalloproteinase 2/metabolism , Intravitreal Injections , Glial Fibrillary Acidic Protein/metabolism , Actins/metabolism , Mice, Inbred C57BL , Recombinant Fusion Proteins
12.
Epigenomics ; 16(14): 985-998, 2024.
Article in English | MEDLINE | ID: mdl-39072366

ABSTRACT

Aim: This study aimed to investigate the role of LINC00513 in colorectal cancer (CRC) progression.Materials & methods: Cell proliferation was evaluated using Cell Counting Kit-8. Cell migration was detected with transwell assay. RNA pull-down was applied for verifying the interactions between LINC00513, IGF2BP1 and connective tissue growth factor (CTGF).Results: LINC00513, IGF2BP1 and CTGF levels were upregulated in CRC. Knockdown of LINC00513 significantly inhibited the malignant behavior of CRC cells. LINC00513 increased CTGF mRNA stability by binding with IGF2BP1. Furthermore, overexpression of IGF2BP1 or CTGF reversed the inhibitory effect of LINC00513 shRNA on CRC progression.Conclusion: LINC00513 promoted CRC cell malignant behaviors through IGF2BP1/CTGF.


Colorectal cancer (CRC) progression seriously threatens the health of people. This study showed that LINC00513 (a long noncoding RNA), insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1) and connective tissue growth factor (CTGF) were significantly upregulated in CRC tissues. Furthermore, the knockdown of LINC00513 inhibited CRC malignant progression in vitro. Mechanistically, LINC00513 increased CTGF mRNA stability in CRC cells by binding with IGF2BP1. As expect, the impact of LINC00513 downregulation on CRC cell proliferation and migration was declined by the overexpression of IGF2BP1 or CTGF. Taken together, LINC00513 upregulation promoted CRC malignant progression by regulating the IGF2BP1/CTGF axis. We believe that this study will help overcome CRC.


Subject(s)
Cell Movement , Cell Proliferation , Colorectal Neoplasms , Connective Tissue Growth Factor , Gene Expression Regulation, Neoplastic , RNA Stability , RNA, Long Noncoding , RNA, Messenger , RNA-Binding Proteins , Humans , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cell Line, Tumor , Disease Progression
13.
Am J Physiol Gastrointest Liver Physiol ; 327(2): G295-G305, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38954823

ABSTRACT

Crohn's disease (CD) is an inflammatory bowel disease characterized by transmural inflammation and intestinal fibrosis. Mechanisms of fibrosis in CD are not well understood. Transmural inflammation is associated with inflammatory cell infiltration, stenosis, and distention, which present mechanical stress (MS) to the bowel wall. We hypothesize that MS induces gene expression of profibrotic mediators such as connective tissue growth factor (CTGF), which may contribute to fibrosis in CD. A rodent model of CD was induced by intracolonic instillation of TNBS to the distal colon. TNBS instillation induced a localized transmural inflammation (site I), with a distended colon segment (site P) proximal to site I. We detected significant fibrosis and collagen content not only in site I but also in site P in CD rats by day 7. CTGF expression increased significantly in sites P and I, but not in the segment distal to the inflammation site. Increased CTGF expression was detected mainly in the smooth muscle cells (SMCs). When rats were fed exclusively with clear liquid diet to prevent mechanical distention in colitis, expression of CTGF in sites P and I was blocked. Direct stretch led to robust expression of CTGF in colonic SMC. Treatment of CD rats with anti-CTGF antibody FG-3149 reduced fibrosis and collagen content in both sites P and I and exhibited consistent trends toward normalizing expression of collagen mRNAs. In conclusion, our studies suggest that mechanical stress, by upregulating profibrotic mediators, i.e., CTGF, may play a critical role in fibrosis in CD.NEW & NOTEWORTHY We found that CTGF expression increased significantly not only in the inflammation site but in the distended segment proximal to inflammation in a rodent model of CD-like colitis. Release of mechanical distention prevented CTGF expression in CD rats, whereas direct stretch induced CTGF expression. Treatment with anti-CTGF antibody reduced fibrosis and collagen contents in CD rats. Thus, mechanical stress, via upregulating profibrotic mediators, i.e., CTGF, may play a critical role in fibrosis in CD.


Subject(s)
Connective Tissue Growth Factor , Crohn Disease , Fibrosis , Rats, Sprague-Dawley , Stress, Mechanical , Animals , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Crohn Disease/metabolism , Crohn Disease/pathology , Rats , Male , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Trinitrobenzenesulfonic Acid , Collagen/metabolism
14.
Otol Neurotol ; 45(7): 810-817, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38995724

ABSTRACT

HYPOTHESIS: Transforming growth factor beta-1 (TGFß-1) and connective tissue growth factor (CTGF) are upregulated in the implanted human cochlea. BACKGROUND: Cochlear implantation can lead to insertion trauma and intracochlear new tissue formation, which can detrimentally affect implant performance. TGFß-1 and CTGF are profibrotic proteins implicated in various pathologic conditions, but little is known about their role in the cochlea. The present study aimed to characterize the expression of these proteins in the human implanted cochlea. METHODS: Archival human temporal bones (HTB) acquired from 12 patients with previous CI and histopathological evidence of new tissue formation as well as surgical samples of human intracochlear scar tissue surrounding the explanted CI were used in this study. Histopathologic analysis of fibrosis and osteoneogenesis was conducted using H&E. Protein expression was characterized using immunofluorescence. RNA expression from surgical specimens of fibrotic tissue surrounding the CI was quantified using qRT-PCR. RESULTS: TGFß-1 and CTGF protein expressions were upregulated in the areas of fibrosis and osteoneogenesis surrounding the CI HTB. Similarly, surgical samples demonstrated upregulation of protein and mRNA expression of TGFß-1 and mild upregulation of CTGF compared with control. TGFß-1 was expressed diffusely within the fibrous capsule, whereas CTGF was expressed in the thickened portion toward the modiolus and the fibrosis-osteoneogensis junction. CONCLUSION: To our knowledge, this is the first study to demonstrate increased expression of TGFß-1 and CTGF in the human implanted cochlea and may provide better understanding of the mechanism behind this pathogenic process to better develop future mitigating interventions.


Subject(s)
Cochlea , Connective Tissue Growth Factor , Transforming Growth Factor beta1 , Humans , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cochlea/metabolism , Male , Middle Aged , Female , Cochlear Implantation , Cochlear Implants , Temporal Bone/metabolism , Temporal Bone/pathology , Fibrosis , Aged , Adult
15.
Clin Oral Investig ; 28(8): 448, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39060456

ABSTRACT

AIM: Resveratrol is a natural polyphenolic compound with biological activities such as anti-inflammation and antioxidation. Its anti-fibrotic effect has been experimentally demonstrated in the pancreas and liver. This study aims to determine the anti-proliferative effect of resveratrol on fibroblasts obtained from hyperplastic gingival tissues from a patient diagnosed with Juvenile Hyaline Fibromatosis (JHF). MATERIALS AND METHODS: Primary gingival fibroblast cell lines were obtained from gingival growth tissues by the gingivectomy of a patient with JHF. Gingival fibroblasts were treated with or without 3 different doses of resveratrol (50, 100, 200 µM). Cytotoxicity and cell proliferation were evaluated after 24, 48, and 72 h. Collagen, TGF, and CTGF were analyzed by ELISA in the 48-hour supernatants. RESULTS: All three doses of resveratrol suppressed the proliferation of JHF gingival fibroblasts at 24 and 48 h without showing any cytotoxic effect compared to the control group (p < 0.0001). At 72 h, 100 and 200 µM resveratrol showed significantly less proliferation (p < 0.0001), less collagen, CTGF, and TGF- ß (p < 0.001) than the control group. CONCLUSION: Resveratrol had a profound anti-proliferative effect on gingival fibroblasts obtained from gingival enlargements with JHF, suggesting that it can be used as a therapeutic to prevent excessive cell growth by suppressing collagen, CTGF, and TGF- ß synthesis in the pathogenesis of hyperplasia.


Subject(s)
Cell Proliferation , Fibroblasts , Resveratrol , Humans , Resveratrol/pharmacology , Fibroblasts/drug effects , Cell Proliferation/drug effects , Gingiva/cytology , Gingiva/drug effects , Enzyme-Linked Immunosorbent Assay , Transforming Growth Factor beta , Collagen , Connective Tissue Growth Factor , Cells, Cultured , Fibromatosis, Gingival/drug therapy , Gingivectomy
16.
Lasers Med Sci ; 39(1): 194, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052077

ABSTRACT

The aim of this study was to evaluate the antiproliferative properties of low-level laser therapy (LLLT) on gingival fibroblasts obtained from calcium channel blocker-induced gingival overgrowth (GO). Gingival fibroblasts of patients with GO were compared to healthy gingival fibroblasts (H). Both cells were exposed to LLLT (685 nm wavelength, 25mW power, diode laser) and compared to those not treated with LLLT. Cell proliferation and viability were measured with MTT assay at baseline and after 24 and 72 h. TGF-ß1, CTGF, and collagen Type 1 levels were evaluated with Enzyme-Linked Immunosorbent Assay (ELISA). LLLT significantly decreased the proliferation of GO fibroblasts (p < 0.05) while leading to a significantly higher proliferation in H fibroblasts compared to the untreated cells (p < 0.05). GO cells showed significantly higher CTGF, TGF-ß, and collagen Type 1 expression than the H cells (p < 0.05). LLLT significantly reduced CTGF levels in GO cells compared to the control group (p < 0.05). In H cells, CTGF and TGF-ß levels were also significantly decreased in response to LLLT compared to the control group (p < 0.05). While LLLT significantly reduced collagen expression in the H group (p < 0.05), it did not significantly impact the GO cells. LLLT significantly reduced the synthesis of the growth factors and collagen in both groups with an antiproliferative effect on the gingival fibroblasts from calcium channel blocker-induced GO, suggesting that it can offer a therapeutic approach in the clinical management of drug-induced GO, reversing the fibrotic changes.


Subject(s)
Calcium Channel Blockers , Cell Proliferation , Connective Tissue Growth Factor , Fibroblasts , Gingiva , Gingival Overgrowth , Low-Level Light Therapy , Humans , Fibroblasts/radiation effects , Fibroblasts/drug effects , Low-Level Light Therapy/methods , Gingival Overgrowth/chemically induced , Gingival Overgrowth/radiotherapy , Gingival Overgrowth/therapy , Calcium Channel Blockers/pharmacology , Cell Proliferation/radiation effects , Cell Proliferation/drug effects , Gingiva/radiation effects , Gingiva/cytology , Connective Tissue Growth Factor/metabolism , Cells, Cultured , Collagen Type I/metabolism , Transforming Growth Factor beta1/metabolism , Cell Survival/radiation effects , Cell Survival/drug effects , Lasers, Semiconductor/therapeutic use , Male , Adult , Female
17.
BMC Med Imaging ; 24(1): 173, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997649

ABSTRACT

BACKGROUND: This study aims to investigate the role of shear wave elastography (SWE) and connective tissue growth factor (CTGF) in the assessment of papillary thyroid carcinoma (PTC) prognosis. METHODS: CTGF expression was detected with immunohistochemistry. Clinical and pathological data were collected. Parameters of conventional ultrasound combined with SWE were also collected. The relationship among CTGF expression, ultrasound indicators, the elastic modulus and the clinicopathological parameters were analyzed. RESULTS: Univariate analysis showed that patients with high risk of PTC were characterized with male, Uygur ethnicity, increased expression of CTGF, convex lesions, calcified, incomplete capsule, intranodular blood flow, rear echo attenuation, cervical lymph node metastasis, lesions larger than 1 cm, psammoma bodies, advanced clinical stage, increased TSH and high value in the shear modulus (P < 0.05). Multivariate analysis demonstrated that the risk factors of high expression of CTGF according to contribution size order were irregular shape, aspect ratio ≥ 1, and increased TSH. The logistic regression model equation was Logit (P) = 1.153 + 1.055 × 1 + 0.926 × 2 + 1.190 × 3 and the Area Under Curve value of the logistic regression was calculated to be 0.850, with a 95% confidence interval of 0.817 to 0.883. CONCLUSION: SWE and CTGF are of great value in the risk assessment of PTC. The degree of fibrosis of PTC is closely related to the prognosis. The hardness of PTC lesions and the expression level of CTGF are correlated with the main indexes of conventional ultrasound differentiating benign or malignant nodules. Irregular shape, aspect ratio ≥ 1, and increased TSH are independent factors of CTGF.


Subject(s)
Connective Tissue Growth Factor , Elasticity Imaging Techniques , Thyroid Cancer, Papillary , Thyroid Neoplasms , Ultrasonography, Doppler, Color , Humans , Male , Elasticity Imaging Techniques/methods , Connective Tissue Growth Factor/metabolism , Female , Thyroid Cancer, Papillary/diagnostic imaging , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Middle Aged , Thyroid Neoplasms/diagnostic imaging , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Risk Assessment , Adult , Prognosis , Aged , Elastic Modulus , Risk Factors
18.
Article in Chinese | MEDLINE | ID: mdl-38964903

ABSTRACT

Objective: To investigate the role of connective tissue growth factor (CTGF) and PI3K/Akt signaling pathways in paraquat (PQ) -induced alterations in alveolar epithelial cell mesenchymalization (EMT) . Methods: In February 2023, RLE-6TN cells were divided into 2 groups, which were set as uncontaminated group and contaminated group (200 µmol/L PQ), and cellular EMT alteration, CTGF and PI3K/Akt signaling pathway related molecules expression were detected by cell scratch assay, qRT-PCR and western-blot assay. Using shRNA interference technology to specifically inhibit the expression of CTGF, RLE-6TN cells were divided into four groups: control group, PQ group (200 µmol/L PQ), interference group (transfected with a plasmid with shRNA-CTGF+200 µmol/L PQ), and null-loaded group (transfected with a plasmid with scramble- CTGF+200 µmol/L PQ), qRT-PCR and western blot were used to examine the alteration of the cellular EMT and the expression of molecules related to the activity of PI3K/Akt pathway. The PI3K/Akt signaling pathway was blocked by the PI3K inhibitor LY294002, and the expression of EMT-related molecules in cells of the control group, PQ group (200 µmol/L PQ), and inhibitor group (200 µmol/L PQ+20 µmol/L LY294002) was examined by qRT-PCR and western blot.The t-test was used to compare the differences between the two groups, while the analysis of variance (ANOVA) was applied to compare the differences among multiple groups. For further pairwise comparisons, the Bonferroni method was adopted. Results: The results of cell scratch test showed that compared with the uncontaminated group, RLE-6TN cells in the contaminated group had faster migration rate, lower mRNA and protein expression levels of E-Cadherin, and higher mRNA and protein expression levels of α-SMA, CTGF, PI3K and Akt, with statistical significance (P<0.05). After specific inhibition of CTGF expression, the mRNA and protein expression of CTGF, PI3K, Akt, and α-SMA in the cells of the interference group were significantly lower than that of the PQ group and the null-loaded group (P<0.05/6), whereas that of E-Cadherin was higher than that of the PQ group and the null-loaded group (P<0.05/6). Specifically blocking the PI3K/Akt signaling pathway, the mRNA and protein expression of PI3K, Akt and α-SMA in the cells of the inhibitor group was decreased compared with that of the PQ group (P<0.05/3), while the expression of E-Cadherin was elevated compared with that of the PQ group (P<0.05/3) . Conclusion: CTGF may promote PQ-induced alveolar epithelial cell EMT through activation of the PI3K/Akt signaling pathway. Inhibition of CTGF expression or blockade of PI3K/Akt signaling pathway activity can alleviate the extent of PQ-induced alveolar epithelial cell EMT.


Subject(s)
Connective Tissue Growth Factor , Epithelial-Mesenchymal Transition , Paraquat , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Connective Tissue Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Epithelial-Mesenchymal Transition/drug effects , Paraquat/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Animals , Rats , Cell Line , Morpholines/pharmacology , Chromones/pharmacology , Cadherins/metabolism
19.
Anticancer Drugs ; 35(8): 709-719, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38900643

ABSTRACT

Glioblastoma (GBM) is a highly angiogenic malignancy of the central nervous system that resists standard antiangiogenic therapy, in part because of an alternative process to angiogenesis termed vasculogenic mimicry. Intricately linked to GBM, dysregulation of the Hippo signaling pathway leads to overexpression of YAP/TEAD and several downstream effectors involved in therapy resistance. Little is known about whether vasculogenic mimicry and the Hippo pathway intersect in the GBM chemoresistance phenotype. This study seeks to investigate the expression patterns of Hippo pathway regulators within clinically annotated GBM samples, examining their involvement in vitro regarding vasculogenic mimicry. In addition, it aims to assess the potential for pharmacological targeting of this pathway. In-silico analysis of the Hippo signaling members YAP1 , TEAD1 , AXL , NF2 , CTGF , and CYR61 transcript levels in low-grade GBM and GBM tumor tissues was done by Gene Expression Profiling Interactive Analysis. Gene expression was analyzed by real-time quantitative PCR from human U87, U118, U138, and U251 brain cancer cell lines and in clinically annotated brain tumor cDNA arrays. Transient gene silencing was performed with specific small interfering RNA. Vasculogenic mimicry was assessed using a Cultrex matrix, and three-dimensional capillary-like structures were analyzed with Wimasis. CYR61 and CTGF transcript levels were elevated in GBM tissues and were further induced when in-vitro vasculogenic mimicry was assessed. Silencing of CYR61 and CTGF , or treatment with a small-molecule TEAD inhibitor LM98 derived from flufenamic acid, inhibited vasculogenic mimicry. Silencing of SNAI1 and FOXC2 also altered vasculogenic mimicry and reduced CYR61 / CTGF levels. Pharmacological targeting of the Hippo pathway inhibits in-vitro vasculogenic mimicry. Unraveling the connections between the Hippo pathway and vasculogenic mimicry may pave the way for innovative therapeutic strategies.


Subject(s)
Brain Neoplasms , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Glioblastoma , Transcription Factors , Humans , Glioblastoma/blood supply , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Connective Tissue Growth Factor/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Transcription Factors/genetics , Neovascularization, Pathologic/drug therapy , TEA Domain Transcription Factors , Gene Expression Regulation, Neoplastic/drug effects , Signal Transduction/drug effects , YAP-Signaling Proteins
20.
Cell Biochem Biophys ; 82(3): 2157-2170, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38837011

ABSTRACT

The CCN family is a group of matricellular proteins associated with the extracellular matrix. This study aims to explore the role of the CCN family in glioma development and its implications in the tumor microenvironment. Through analysis of bulk RNA-seq cohorts, correlations between CCN family expression and glioma subtypes, patient survival, and bioactive pathway enrichment were investigated. Additionally, single-cell datasets were employed to identify novel cell subgroups, followed by analyses of cell communication and transcription factors. Spatial transcriptomic analysis was utilized to validate the CCN family's involvement in glioma. Results indicate overexpression of CYR61,CTGF, and WISP1 in glioma, associated with unfavorable subtypes and reduced survival. Enrichment analyses revealed associations with oncogenic pathways, while CTGF and WISP1 expression correlated with increased infiltration of regulatory T cells and M2 macrophages. Single-cell analysis identified MES-like cells as the highest CCN expression. Moreover, intercellular signal transduction analysis demonstrated active pathways, including SPP1-CD44, in cell subgroups with elevated CYR61 and CTGF expression. Spatial transcriptomic analysis confirmed co-localization of CYR61,CTGF and SPP1-CD44 with high oncogenic pathway activity. These findings suggest that CCN family members may serve as potential prognostic biomarkers and therapeutic targets for glioma.


Subject(s)
CCN Intercellular Signaling Proteins , Extracellular Matrix , Glioma , Humans , Glioma/metabolism , Glioma/pathology , Glioma/genetics , Prognosis , Extracellular Matrix/metabolism , CCN Intercellular Signaling Proteins/metabolism , CCN Intercellular Signaling Proteins/genetics , Tumor Microenvironment , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Single-Cell Analysis , Cysteine-Rich Protein 61/metabolism , Cysteine-Rich Protein 61/genetics , Tumor Escape , Gene Expression Regulation, Neoplastic , Gene Expression Profiling , Signal Transduction , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Multiomics
SELECTION OF CITATIONS
SEARCH DETAIL