Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
1.
Int J Mol Med ; 54(5)2024 Nov.
Article in English | MEDLINE | ID: mdl-39301627

ABSTRACT

The present study aimed to investigate the role and mechanism of inhibin ßA (INHBA) in thyroid cancer (TC), and to determine its potential impact on the aggressive behavior of TC cells. The present study employed a comprehensive approach, using public databases, such as the Gene Expression Omnibus and The Cancer Genome Atlas, to identify and analyze the expression of INHBA in TC. Cell transfection, reverse transcription­quantitative PCR, western blot analysis, immunohistochemistry and in vivo assays were conducted to investigate the functional effects of INHBA on TC. In addition, the present study explored the molecular mechanisms underlying the effects of INHBA, focusing on the potential impact on the RhoA signaling pathway and associated molecular cascades. Bioinformatics analysis revealed a significant association between INHBA expression and TC, and INHBA expression was markedly upregulated in TC tissues compared with in healthy control tissues. The results of functional studies demonstrated that INHBA overexpression increased the migration and invasion of TC cells, and the opposite result was observed following INHBA knockdown. Mechanistic investigations indicated that INHBA modulated the RhoA pathway, leading to alterations in the phosphorylation status of LIM kinase 1 (LIMK) and cofilin, key regulators of cytoskeletal dynamics and cell motility. Following the introduction of transfected TC cells into zebrafish and nude mouse models, the results of the present study demonstrated that INHBA knockdown attenuated the metastatic potential of TC cells. In conclusion, INHBA may serve a pivotal role in promoting the aggressive phenotype of TC cells through modulating the RhoA/LIMK/cofilin signaling axis. These findings highlight INHBA as a potential biomarker and therapeutic target for the management of aggressive TC.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Inhibin-beta Subunits , Thyroid Neoplasms , rhoA GTP-Binding Protein , Humans , Animals , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Cell Line, Tumor , rhoA GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , Cell Movement/genetics , Mice , Neoplasm Metastasis , Signal Transduction , Mice, Nude , Female , Male , Zebrafish , Lim Kinases/metabolism , Lim Kinases/genetics , Middle Aged , Actin Depolymerizing Factors/metabolism , Actin Depolymerizing Factors/genetics
2.
Cancer Med ; 13(17): e7368, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39248018

ABSTRACT

OBJECTIVE: The TGF-ß superfamily member activin, a dimer of the gene products of INHBA and/or INHBB, has been implicated in immune cell maturation and recruitment, but its immune impact within epithelial ovarian cancer (EOC) is not well characterized. We sought to explore differences in activin (INHBA/ Inhibin-ßA and INHBB/ Inhibin-ßB) between malignant and ovarian tissues at the RNA and protein level and assess the relationship between activin and immune cells in EOC. METHODS: Publicly available RNA sequencing data were accessed from GEO (#GSE143897) with normalization and quantification performed via DESeq2. Immune gene expression profile was further explored within the TCGA-OV cohort derived from The Cancer Genome Atlas (TCGA). Immunohistochemical analysis was performed to evaluate activin A and T-cell markers CD8 and FoxP3 at the protein level. ELISA to activin-A was used to assess levels in the ascites of advanced EOC patients. Kaplan-Meier curves were generated to visualize survival outcomes. RESULTS: Gene expression levels of components of the activin signaling pathway were elevated within EOC when compared to a benign cohort, with differences in activin type I/II receptor gene profiles identified. Additionally, INHBA gene expression was linked to lymphocytic immune markers in EOC samples. Immunohistochemistry analysis revealed a positive correlation of CD8 and FOXP3 staining with activin A at the protein level in both primary and metastatic epithelial ovarian cancer samples. Furthermore, Activin-A (inhibin-ßA) is significantly elevated in EOC patient ascites. CONCLUSION: INHBA expression is elevated within EOC, correlating with worse survival, with activin protein levels correlating with specific immune infiltration. Our findings suggest that activin-A may play a role in suppressing anti-tumor immunity in EOC, highlighting its potential as a therapeutic target.


Subject(s)
Activins , Carcinoma, Ovarian Epithelial , Inhibin-beta Subunits , Ovarian Neoplasms , Humans , Female , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Ovarian Neoplasms/genetics , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Activins/metabolism , Activins/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Middle Aged , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics
3.
Sci Rep ; 14(1): 14625, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38918475

ABSTRACT

Visfatin (VIS) is a hormone belonging to the adipokines' group secreted mainly by the adipose tissue. VIS plays a crucial role in the control of energy homeostasis, inflammation, cell differentiation, and angiogenesis. VIS expression was confirmed in the hypothalamic-pituitary-gonadal (HPG) axis structures, as well as in the uterus, placenta, and conceptuses. We hypothesised that VIS may affect the abundance of proteins involved in the regulation of key processes occurring in the corpus luteum (CL) during the implantation process in pigs. In the present study, we performed the high-throughput proteomic analysis (liquid chromatography with tandem mass spectrometry, LC-MS/MS) to examine the in vitro influence of VIS (100 ng/mL) on differentially regulated proteins (DRPs) in the porcine luteal cells (LCs) on days 15-16 of pregnancy (implantation period). We have identified 511 DRPs, 276 of them were up-regulated, and 235 down-regulated in the presence of VIS. Revealed DRPs were assigned to 162 gene ontology terms. Western blot analysis of five chosen DRPs, ADAM metallopeptidase with thrombospondin type 1 motif 1 (ADAMTS1), lanosterol 14-α demethylase (CYP51A1), inhibin subunit beta A (INHBA), notch receptor 3 (NOTCH3), and prostaglandin E synthase 2 (mPGES2) confirmed the veracity and accuracy of LC-MS/MS method. We indicated that VIS modulates the expression of proteins connected with the regulation of lipogenesis and cholesterologenesis, and, in consequence, may be involved in the synthesis of steroid hormones, as well as prostaglandins' metabolism. Moreover, we revealed that VIS affects the abundance of protein associated with ovarian cell proliferation, differentiation, and apoptosis, as well as CL new vessel formation and tissue remodelling. Our results suggest important roles for VIS in the regulation of ovarian functions during the peri-implantation period.


Subject(s)
Embryo Implantation , Luteal Cells , Nicotinamide Phosphoribosyltransferase , Proteome , Animals , Female , Swine , Nicotinamide Phosphoribosyltransferase/metabolism , Proteome/metabolism , Luteal Cells/metabolism , Pregnancy , Proteomics/methods , Tandem Mass Spectrometry , Chromatography, Liquid , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/genetics
4.
Medicine (Baltimore) ; 103(23): e38416, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847724

ABSTRACT

To investigate the expression of Inhibin B between various clinical stages, Chinese medicine dialectic typing, and in nasopharyngeal carcinoma (NPC) tissues and serum, and to evaluate the potential of Inhibin B as a new biomarker for NPC. Paraffin specimens of pathologically confirmed NPC tissues and paracancerous tissues were retrospectively collected, and the expression of Inhibin α (INHA) and Inhibin ßB (INHBB) was detected by SP method, and their relationship with clinicopathological indexes was analyzed; in addition, patients with NPC who had received radiotherapy were included as the study subjects, and Epstein-Barr virus DNA (EBV-DNA), INHA, and INHBB in patients were detected by using the fluorescence quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and chemiluminescent immuno-sandwiching method, respectively. EBV-DNA, EBV-viral capsid antigen-immunoglobulin A (VCA IgA), INHA, and INHBB were detected in the patients, respectively, and their relationships with traditional Chinese medicine (TCM) patterns were also analyzed. The expression of INHA and INHBB in NPC tissues was lower than that in paracancerous tissues, and the expression of INHA in NPC patients was correlated with lymphatic metastasis, clinical staging, and TCM staging; the levels of EBV-DNA and VCA IgA were higher than that of healthy populations in NPC patients and were higher than that of patients with stage III + IV than that of patients with stage I + II, and the levels of INHA and INHBB were lower than those of healthy populations and were lower than those of patients with stage III + IV than that of patients with stage I + II. The levels of INHA and INHBB in nasopharyngeal cancer patients were lower than those in healthy people, and the levels in stage III + IV patients were lower than those in stage I + II patients. The levels of EBV-DNA and VCA IgA in nasopharyngeal cancer patients were correlated with the Chinese medicine patterns, and had different patterns. The expression of Inhibin B may be related to the progression of NPC, and it has certain typing significance for different TCM syndromes of NPC, which is helpful for TCM typing diagnosis.


Subject(s)
Medicine, Chinese Traditional , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Humans , Male , Female , Nasopharyngeal Neoplasms/virology , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/blood , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Carcinoma/diagnosis , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Carcinoma/blood , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Medicine, Chinese Traditional/methods , Middle Aged , Retrospective Studies , Adult , DNA, Viral/analysis , DNA, Viral/blood , Inhibins/blood , Herpesvirus 4, Human/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/blood , Neoplasm Staging , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/blood , Aged , Antigens, Viral/blood , Immunoglobulin A/blood , Capsid Proteins
5.
Int J Biol Macromol ; 270(Pt 1): 132239, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735606

ABSTRACT

Colorectal cancer (CRC) is a major worldwide health issue, with high rates of both occurrence and mortality. Dysregulation of the transforming growth factor-beta (TGF-ß) signaling pathway is recognized as a pivotal factor in CRC pathogenesis. Notably, the INHBA gene and long non-coding RNAs (lncRNAs) have emerged as key contributors to CRC progression. The aim of this research is to explore the immunological roles of INHBA and PELATON in CRC through a combination of computational predictions and experimental validations, with the goal of enhancing diagnostic and therapeutic strategies. In this study, we utilized bioinformatics analyses, which involved examining differential gene expression (DEG) in the TCGA-COAD dataset and exploring the INHBA gene in relation to the TGF-ß pathway. Additionally, we analyzed mutations of INHBA, evaluated the microenvironment and tumor purity, investigated the INHBA's connection to immune checkpoint inhibitors, and measured its potential as an immunotherapy target using the TIDE score. Utilizing bioinformatics analyses of the TCGA-COAD dataset beside experimental methodologies such as RT-qPCR, our investigation revealed significant upregulation of INHBA in CRC. As results, our analysis of the protein-protein interaction network associated with INHBA showed 10 interacting proteins that play a role in CRC-associated processes. We observed a notable prevalence of mutations within INHBA and explored its correlation with the response to immune checkpoint inhibitors. Our study highlights INHBA as a promising target for immunotherapy in CRC. Moreover, our study identified PELATON as a closely correlated lncRNA with INHBA, with experimental validation confirming their concurrent upregulation in CRC tissues. Thus, these findings highlight the importance of INHBA and PELATON in driving CRC progression, suggesting their potential utility as diagnostic and prognostic biomarkers. By integrating computational predictions with experimental validations, this research enhances our understanding of CRC pathogenesis and uncovers prospects for personalized therapeutic interventions.


Subject(s)
Colorectal Neoplasms , Computational Biology , Gene Expression Regulation, Neoplastic , Inhibin-beta Subunits , Protein Interaction Maps , Signal Transduction , Transforming Growth Factor beta , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Humans , Computational Biology/methods , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Protein Interaction Maps/genetics , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , RNA, Long Noncoding/genetics , Tumor Microenvironment/genetics , Mutation , Biomarkers, Tumor/genetics
6.
Mol Metab ; 78: 101830, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38787338

ABSTRACT

OBJECTIVE: The liver is a central regulator of energy metabolism exerting its influence both through intrinsic processing of substrates such as glucose and fatty acid as well as by secreting endocrine factors, known as hepatokines, which influence metabolism in peripheral tissues. Human genome wide association studies indicate that a predicted loss-of-function variant in the Inhibin ßE gene (INHBE), encoding the putative hepatokine Activin E, is associated with reduced abdominal fat mass and cardiometabolic disease risk. However, the regulation of hepatic Activin E and the influence of Activin E on adiposity and metabolic disease are not well understood. Here, we examine the relationship between hepatic Activin E and adipose metabolism, testing the hypothesis that Activin E functions as part of a liver-adipose, inter-organ feedback loop to suppress adipose tissue lipolysis in response to elevated serum fatty acids and hepatic fatty acid exposure. METHODS: The relationship between hepatic Activin E and non-esterified fatty acids (NEFA) released from adipose lipolysis was assessed in vivo using fasted CL 316,243 treated mice and in vitro using Huh7 hepatocytes treated with fatty acids. The influence of Activin E on adipose lipolysis was examined using a combination of Inhbe knockout mice, a mouse model of hepatocyte-specific overexpression of Activin E, and mouse brown adipocytes treated with Activin E enriched media. RESULTS: Increasing hepatocyte NEFA exposure in vivo by inducing adipose lipolysis through fasting or CL 316,243 treatment increased hepatic Inhbe expression. Similarly, incubation of Huh7 human hepatocytes with fatty acids increased expression of INHBE. Genetic ablation of Inhbe in mice increased fasting circulating NEFA and hepatic triglyceride accumulation. Treatment of mouse brown adipocytes with Activin E conditioned media and overexpression of Activin E in mice suppressed adipose lipolysis and reduced serum FFA levels, respectively. The suppressive effects of Activin E on lipolysis were lost in CRISPR-mediated ALK7 deficient cells and ALK7 kinase deficient mice. Disruption of the Activin E-ALK7 signaling axis in Inhbe KO mice reduced adiposity upon HFD feeding, but caused hepatic steatosis and insulin resistance. CONCLUSIONS: Taken together, our data suggest that Activin E functions as part of a liver-adipose feedback loop, such that in response to increased serum free fatty acids and elevated hepatic triglyceride, Activin E is released from hepatocytes and signals in adipose through ALK7 to suppress lipolysis, thereby reducing free fatty acid efflux to the liver and preventing excessive hepatic lipid accumulation. We find that disrupting this Activin E-ALK7 inter-organ communication network by ablation of Inhbe in mice increases lipolysis and reduces adiposity, but results in elevated hepatic triglyceride and impaired insulin sensitivity. These results highlight the liver-adipose, Activin E-ALK7 signaling axis as a critical regulator of metabolic homeostasis.


Subject(s)
Activins , Adipose Tissue , Fatty Acids , Inhibin-beta Subunits , Lipolysis , Liver , Animals , Humans , Mice , Activins/metabolism , Adipose Tissue/metabolism , Adiposity , Fatty Acids/metabolism , Fatty Acids, Nonesterified/metabolism , Fatty Acids, Nonesterified/blood , Hepatocytes/metabolism , Inhibin-beta Subunits/metabolism , Inhibin-beta Subunits/genetics , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout
7.
PLoS Genet ; 18(12): e1010523, 2022 12.
Article in English | MEDLINE | ID: mdl-36469526

ABSTRACT

Activin and inhibin are both dimeric proteins sharing the same ß subunits that belong to the TGF-ß superfamily. They are well known for stimulating and inhibiting pituitary FSH secretion, respectively, in mammals. In addition, activin also acts as a mesoderm-inducing factor in frogs. However, their functions in development and reproduction of other species are poorly defined. In this study, we disrupted all three activin/inhibin ß subunits (ßAa, inhbaa; ßAb, inhbab; and ßB, inhbb) in zebrafish using CRISPR/Cas9. The loss of ßAa/b but not ßB led to a high mortality rate in the post-hatching stage. Surprisingly, the expression of fshb but not lhb in the pituitary increased in the female ßA mutant together with aromatase (cyp19a1a) in the ovary. The single mutant of ßAa/b showed normal folliculogenesis in young females; however, their double mutant (inhbaa-/-;inhbab-/-) showed delayed follicle activation, granulosa cell hypertrophy, stromal cell accumulation and tissue fibrosis. The ovary of inhbaa-/- deteriorated progressively after 180 dpf with reduced fecundity and the folliculogenesis ceased completely around 540 dpf. In addition, tumor- or cyst-like tissues started to appear in the inhbaa-/- ovary after about one year. In contrast to females, activin ßAa/b mutant males showed normal spermatogenesis and fertility. As for activin ßB subunit, the inhbb-/- mutant exhibited normal folliculogenesis, spermatogenesis and fertility in both sexes; however, the fecundity of mutant females decreased dramatically at 270 dpf with accumulation of early follicles. In summary, the activin-inhibin system plays an indispensable role in fish reproduction, in particular folliculogenesis and ovarian homeostasis.


Subject(s)
Inhibin-beta Subunits , Inhibins , Animals , Female , Inhibins/genetics , Inhibins/metabolism , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Activins/genetics , Activins/metabolism , Reproduction/genetics , Mammals/metabolism
8.
Front Endocrinol (Lausanne) ; 13: 919670, 2022.
Article in English | MEDLINE | ID: mdl-35909548

ABSTRACT

The Sertoli cells of the testes play an essential role during gonadal development, in addition to supporting subsequent germ cell survival and spermatogenesis. Anti-Müllerian hormone (AMH) is a member of the TGF-ß superfamily, which is secreted by immature Sertoli cells from the 8th week of fetal gestation. lnhibin B is a glycoprotein, which is produced by the Sertoli cells from early in fetal development. In people with a Difference or Disorder of Sex Development (DSD), these hormones may be useful to determine the presence of testicular tissue and potential for spermatogenesis. However, fetal Sertoli cell development and function is often dysregulated in DSD conditions and altered production of Sertoli cell hormones may be detected throughout the life course in these individuals. As such this review will consider the role of AMH and inhibin B in individuals with DSD.


Subject(s)
Anti-Mullerian Hormone , Disorders of Sex Development , Inhibins , Sertoli Cells , Sex Differentiation , Anti-Mullerian Hormone/genetics , Anti-Mullerian Hormone/metabolism , Disorders of Sex Development/genetics , Disorders of Sex Development/metabolism , Humans , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Inhibins/genetics , Inhibins/metabolism , Male , Sertoli Cells/metabolism , Sex Differentiation/physiology , Spermatogenesis/physiology , Testis/metabolism
9.
Sci Rep ; 12(1): 1659, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102236

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) patients display distinct phenotypes of cachexia development, with either adipose tissue loss preceding skeletal muscle wasting or loss of only adipose tissue. Activin A levels were measured in serum and analyzed in tumor specimens of both a cohort of Stage IV PDAC patients and the genetically engineered KPC mouse model. Our data revealed that serum activin A levels were significantly elevated in Stage IV PDAC patients in comparison to age-matched non-cancer patients. Little is known about the role of activin A in adipose tissue wasting in the setting of PDAC cancer cachexia. We established a correlation between elevated activin A and remodeling of visceral adipose tissue. Atrophy and fibrosis of visceral adipose tissue was examined in omental adipose tissue of Stage IV PDAC patients and gonadal adipose tissue of an orthotopic mouse model of PDAC. Remarkably, white visceral adipose tissue from both PDAC patients and mice exhibited decreased adipocyte diameter and increased fibrotic deposition. Strikingly, expression of thermogenic marker UCP1 in visceral adipose tissues of PDAC patients and mice remained unchanged. Thus, we propose that activin A signaling could be relevant to the acceleration of visceral adipose tissue wasting in PDAC-associated cachexia.


Subject(s)
Activins/metabolism , Adipocytes, White/metabolism , Adiposity , Carcinoma, Pancreatic Ductal/metabolism , Inhibin-beta Subunits/metabolism , Intra-Abdominal Fat/metabolism , Pancreatic Neoplasms/metabolism , Activins/genetics , Adipocytes, White/pathology , Animals , Atrophy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Cell Line , Fibrosis , Humans , Inhibin-beta Subunits/genetics , Intra-Abdominal Fat/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Signal Transduction , Uncoupling Protein 1/metabolism
10.
J Cell Mol Med ; 26(5): 1540-1555, 2022 03.
Article in English | MEDLINE | ID: mdl-35150061

ABSTRACT

Pancreatic cancer is one of the most lethal gastrointestinal tumours, the most common pathological type is pancreatic adenocarcinoma (PAAD). In recent year, immune imbalanced in tumour microenvironment has been shown to play an important role in the evolution of tumours progression, and the efficacy of immunotherapy has been gradually demonstrated in clinical practice. In this study, we propose to construct an immune-related prognostic risk model based on immune-related genes MMP14 and INHBA expression that can assess the prognosis of pancreatic cancer patients and identify potential therapeutic targets for pancreatic cancer, to provide new ideas for the treatment of pancreatic cancer. We also investigate the correlation between macrophage infiltration and MMP14 and INHBA expression. First, the gene expression data of pancreatic cancer and normal pancreatic tissue were obtained from The Cancer Genome Atlas Program (TCGA) and The Genotype-Tissue Expression public database (GTEx). The differentially expressed immune-related genes between pancreatic cancer samples and normal sample were screened by R software. Secondly, univariate Cox regression analysis were used to evaluate the relationship between immune-related genes and the prognosis of pancreatic cancer patients. A polygenic risk score model was constructed by Cox regression analysis. The prognostic nomogram was constructed, and its performance was evaluated comprehensively by internal calibration curve and C-index. Using the risk model, each patient gets a risk score, and was divided into high- or low- risk groups. The proportion of 22 types of immune cells infiltration in pancreatic cancer samples was inferred by CIBERSOFT algorithm, correlation analysis (Pearson method) was used to analyse the correlation between the immune-related genes and immunes cells. Then, we applied macrophage conditioned medium to culture pancreatic cancer cell line PANC1, detected the expression of MMP14 and INHBA by qRT-PCR and Western blot methods. Knock-down MMP14 and INHBA in PANC1 cells by transfected with shRNA lentiviruses. Detection of migration ability of pancreatic cells was done by trans-well cell migration assay. A subcutaneous xenograft tumour model of human pancreatic cancer in nude mice was constructed. In conclusion, an immune-related gene prognostic model was constructed, patients with high-risk scores have poorer survival status, M2-phenotype tumour-associated macrophages (TAMs) up-regulate two immune-related genes, MMP14 and INHBA, which were used to establish the prognostic model. Knock-down of MMP14 and INHBA inhibited invasion of pancreatic cancer.


Subject(s)
Adenocarcinoma , Inhibin-beta Subunits/metabolism , Pancreatic Neoplasms , Adenocarcinoma/genetics , Animals , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Phenotype , Prognosis , Tumor Microenvironment/genetics , Tumor-Associated Macrophages , Pancreatic Neoplasms
11.
J Pathol ; 256(1): 25-37, 2022 01.
Article in English | MEDLINE | ID: mdl-34543458

ABSTRACT

Upstream stimuli for myofibroblast activation are of considerable interest for understanding the mechanisms underlying renal fibrosis. Activin B, a member of the TGF-ß family, exists as a homodimer of inhibin subunit beta B (INHBB), but its role in renal fibrosis remains unknown. We found that INHBB expression was significantly increased in various renal fibrosis models and human chronic kidney disease specimens with renal fibrosis. Notably, the increase of INHBB occurred mainly in the tubular epithelial cells (TECs). In vivo, inhibiting INHBB blocked the activation of interstitial fibroblasts and ameliorated the renal fibrosis induced by unilateral ureteral obstruction or ischemia-reperfusion injury, while ectopic expression of INHBB in the TECs was able to activate interstitial fibroblasts and initiate interstitial fibrosis. In vitro, overexpression of INHBB in TECs led to the secretion of activin B, thereby promoting the proliferation and activation of interstitial fibroblasts through activin B/Smad signaling. Furthermore, inhibition of activin B/Smad signaling attenuated the fibrotic response caused by tubular INHBB. Mechanistically, the upregulation of INHBB depended on the transcription factor Sox9 in the injured TECs. Clinical analyses also identified a positive correlation between Sox9 and INHBB expression in human specimens, suggesting the Sox9/INHBB axis as a positive regulator of renal fibrosis. In conclusion, tubule-derived INHBB is implicated in the pathogenesis of renal fibrosis by activating the surrounding fibroblasts in a paracrine manner, thereby exhibiting as a potential therapeutic target. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Fibroblasts/metabolism , Fibrosis/metabolism , Inhibin-beta Subunits/metabolism , Animals , Cell Proliferation/physiology , Fibroblasts/pathology , Fibrosis/pathology , Humans , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Myofibroblasts/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Up-Regulation , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
12.
Pathol Oncol Res ; 27: 1610032, 2021.
Article in English | MEDLINE | ID: mdl-34867090

ABSTRACT

This study explored the roles of activins and follistatin in colorectal cancers. Paired malignant and normal colonic tissues were collected from archived paraffin-embedded (n = 90 patients) alongside fresh (n = 40 patients) specimen cohorts. Activin ß-subunits, follistatin and Smad4 mRNAs and proteins were measured by real-time PCR and immunohistochemistry (IHC). Mature activin-A, -B, -AB and follistatin proteins were measured by ELISA. Cancer tissues having ≤ the 20th percentile of the Smad4 IHC score were considered as low (L-S4) group. The Smad4-intact SW480 and Smad4-null HT29 colon cancer cell lines were treated with activins and follistatin, and cell cycle was analysed by flow cytometry. The cell cycle inducing (CCND1/CCND3) and inhibitory (p21/p27) proteins alongside the survival (survivin/BCL2) and pro-apoptosis (Casp-8/Casp-3) markers were measured by immunofluorescence. Thirty-nine patients had right-sided cancers (30%) and showed higher rates of L-S4 tumours (n = 17; 13.1%) alongside worse clinicopathological characteristics relative to left-sided cancers. The ßA-subunit and activin-A increased, whilst ßB-subunit and activin-AB decreased, in malignant sites and the late-stage cancers revealed the greatest abnormalities. Interestingly, follistatin declined markedly in early-stage malignant tissues, whilst increased significantly in the advanced stages. All activin molecules were comparable between the early stage right- and left-sided tumours, whereas the late-stage right-sided cancers and L-S4 tumours showed more profound deregulations. In vitro, activin-A increased the numbers of the SW480 cells in sub-G1 and G0/G1-phases, whereas reduced the HT29 cell numbers in the sub-G1 phase with simultaneous increases in the G0/G1 and S phases. The p21/p27/Casp-8/Casp-3 proteins escalated, whilst CCND1/CCND3/BCL2/survivin declined in the SW480 cells following activin-A, whereas activin-A only promoted p21 and p27 alongside reduced CCND3 in the HT29 cells. By contrast, activin-AB increased the numbers of SW480 and HT29 cells in Sub-G1 and G0/G1-phases and promoted the anti-cancer and reduced the oncogenic proteins in both cell lines. In conclusion, activins and follistatin displayed stage-dependent dysregulations and were markedly altered during the advanced stages of right-sided and L-S4 cancers. Moreover, the activin-A actions in CRC could be Smad4-dependent, whereas activin-AB may act as a Smad4-independent tumour suppressor protein.


Subject(s)
Activins/metabolism , Colorectal Neoplasms/metabolism , Follistatin/metabolism , Smad4 Protein/metabolism , Activins/genetics , Activins/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Progression , Female , Follistatin/genetics , Follistatin/pharmacology , Humans , Inhibin-beta Subunits/metabolism , Male , RNA, Messenger/genetics , Smad4 Protein/genetics
13.
Bioengineered ; 12(2): 11567-11575, 2021 12.
Article in English | MEDLINE | ID: mdl-34889158

ABSTRACT

Emerging evidence has demonstrated that inhibin subunit beta A (INHBA) is dysregulated and plays a critical role in various cancers. With the development of sequencing technology, studies have discovered that INHBA is overexpressed in breast cancer tissues. However, the biological roles of INHBA in breast cancer are still far to clear. In the present study, we analyzed the INHBA expression in the Cancer Genome Atlas (TCGA) database. Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to assess the expression of INHBA in breast cancer cell lines. Cell proliferation, invasion and epithelial-mesenchymal transition (EMT) were determined by using CCK-8, EdU, Transwell and western blot assays. The result showed that INHBA was highly expressed in breast cancer cell lines. Functional analysis revealed that silence or elevation of INHBA inhibited or promoted the proliferation, migration, invasion and EMT and Wnt/ß-catenin signaling pathway-related markers of MCF-7 cells. Mechanically, blocking of Wnt/ß-catenin pathway by XAV939 reversed the promotion effect of INHBA overexpression on breast cancer cells' proliferation, migration and invasion. Our findings emphasized that INHBA may act as an oncogene via activating the Wnt/ß-catenin pathway, which may provide a potential therapeutic target for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Inhibin-beta Subunits/metabolism , Wnt Signaling Pathway , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Up-Regulation/genetics
14.
J Biol Chem ; 297(4): 101162, 2021 10.
Article in English | MEDLINE | ID: mdl-34481843

ABSTRACT

Cyclin-dependent kinase 7 (CDK7) is a master regulatory kinase that drives cell cycle progression and stimulates expression of oncogenes in a myriad of cancers. Inhibitors of CDK7 (CDK7i) are currently in clinical trials; however, as with many cancer therapies, patients will most likely experience recurrent disease due to acquired resistance. Identifying targets underlying CDK7i resistance will facilitate prospective development of new therapies that can circumvent such resistance. Here we utilized triple-negative breast cancer as a model to discern mechanisms of resistance as it has been previously shown to be highly responsive to CDK7 inhibitors. After generating cell lines with acquired resistance, high-throughput RNA sequencing revealed significant upregulation of genes associated with efflux pumps and transforming growth factor-beta (TGF-ß) signaling pathways. Genetic silencing or pharmacological inhibition of ABCG2, an efflux pump associated with multidrug resistance, resensitized resistant cells to CDK7i, indicating a reliance on these transporters. Expression of activin A (INHBA), a member of the TGF-ß family of ligands, was also induced, whereas its intrinsic inhibitor, follistatin (FST), was repressed. In resistant cells, increased phosphorylation of SMAD3, a downstream mediator, confirmed an increase in activin signaling, and phosphorylated SMAD3 directly bound the ABCG2 promoter regulatory region. Finally, pharmacological inhibition of TGF-ß/activin receptors or genetic silencing of SMAD4, a transcriptional partner of SMAD3, reversed the upregulation of ABCG2 in resistant cells and phenocopied ABCG2 inhibition. This study reveals that inhibiting the TGF-ß/Activin-ABCG2 pathway is a potential avenue for preventing or overcoming resistance to CDK7 inhibitors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/biosynthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Inhibin-beta Subunits/metabolism , Neoplasm Proteins/biosynthesis , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Triple Negative Breast Neoplasms/metabolism , Up-Regulation/drug effects , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cell Line, Tumor , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Drug Resistance, Neoplasm/genetics , Female , Humans , Inhibin-beta Subunits/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Cyclin-Dependent Kinase-Activating Kinase
15.
Mol Med Rep ; 24(5)2021 11.
Article in English | MEDLINE | ID: mdl-34505633

ABSTRACT

Inhibin ßA (INHBA) serves a prognostic and tumor­promoting role in numerous types of cancer. The present study aimed to determine the clinical significance of INHBA in non­small cell lung cancer (NSCLC) and the mechanisms underlying its potential tumor­promoting effect. INHBA expression was detected in clinical NSCLC samples using immunohistochemistry. In vivo loss­ and gain­of­function studies were performed to determine the effects of INHBA on NSCLC invasion. In addition, protein and mRNA expression levels of INHBA, yes­associated protein (YAP), large tumor suppressor 1/2 kinase (LATS1/2), connective tissue growth factor, cysteine rich angiogenic inducer 61 and Merlin were assessed using western blotting and reverse transcription­quantitative PCR, respectively, to investigate the mechanism by which INHBA may affect the invasion of NSCLC. The present study revealed that INHBA was significantly upregulated in 238 clinical NSCLC samples compared with its expression levels in paired adjacent non­cancerous tissues, and in metastatic nodules compared with in primary tumors. Notably, high INHBA expression was statistically associated with clinicopathological features, including poor differentiation and advanced tumor stage. INHBA positivity was statistically related to decreased 5­year overall survival, for which INHBA was an independent prognostic factor. Furthermore, INHBA promoted NSCLC invasion in vitro. In NSCLC, INHBA expression was associated with the nuclear levels of YAP and INHBA overexpression enhanced the invasive abilities of NSCLC cells via inhibiting the Hippo pathway. Mechanistically, INHBA inhibited l LATS1/2 phosphorylation and induced YAP nuclear translocation by downregulating the protein expression levels of Merlin. In conclusion, INHBA may negatively regulate the Hippo pathway to act as a tumor promotor, and could represent a marker of prognosis in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Inhibin-beta Subunits/metabolism , Adult , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , China , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Hippo Signaling Pathway/genetics , Humans , Inhibin-beta Subunits/analysis , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prognosis , Protein Serine-Threonine Kinases , Signal Transduction/drug effects , Transcription Factors/genetics
16.
Bioengineered ; 12(1): 4681-4696, 2021 12.
Article in English | MEDLINE | ID: mdl-34346300

ABSTRACT

Accumulating evidence indicates that INHBA (Inhibin ß-A, a member of the TGF-ß superfamily) functions as an oncogene in cancer progression. However, little is known as to how INHBA regulates the progression and aggressiveness of breast cancer (BC). This study explored the function and underlying mechanism of INHBA in epithelial-mesenchymal transition (EMT) of BC cells. INHBA expression in BC cell lines was measured using RT-qPCR and Western blot. The would-healing and transwell migration assays were used to investigate the effect of INHBA overexpression or silencing on BC cell motility. Moreover, the expression levels of EMT-related genes were quantified after overexpressing or silencing of INHBA. Based on published dataset, INHBA was significantly upregulated in BC tissues compared to the adjacent normal tissues. A higher level of INHBA expression was also correlated with a poor survival in BC patients. In addition, in vitro study showed that INHBA played an indispensable role in promoting BC cell proliferation and invasion. Mechanistically, INHBA induced epithelial-mesenchymal transition (EMT) and accelerated the motility of BC cells by activating TGF-ß-regulated genes. In conclusion, INHBA plays a functional role in supporting EMT phenotype of BC cells, and it may serve as a diagnostic biomarker and a potential therapeutic target for BC treatment.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition/genetics , Inhibin-beta Subunits , Transforming Growth Factor beta , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Mice , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
17.
Gen Comp Endocrinol ; 312: 113856, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34302847

ABSTRACT

Inhibin and Activin, belong to the transforming growth factor ß superfamily (TGF-ß), which associate with the regulation of the reproductive process by the modulation of the hypothalamic-pituitary-gonad (HPG) axis. In this study, we reported the molecular cloning and tissue expression of inhibin α in allotriploid crucian carp and its parent- diploid red crucian carp. The full-length cDNA of inhibin α were respectively 1632 bp and 1642 bp in allotriploids and diploids, which both consisted of a 1044 bp open reading frame (ORF) encoding 347 amino acids. Real-time quantitative PCR (RT-qPCR) showed that allotriploids and diploids had significant expression of inhibin α in testis and ovary, and the expression of inhibin α in the gonads of allotriploids was higher than that of diploids. The immunohistochemistry indicated that the ovarian development of allotriploids was abnormal, and the expression of Inhibin α in the ovary of allotriploids was higher than that of diploids. Results of co-immunoprecitation (co-IP) demonstrated that the Inhibin α and Activin ßA, Inhibin α and Activin ßB can form dimers. These findings suggested that the elevated expression of inhibin α and the competitive binding of Inhibin α subunit with Activin ß subunits in allotriploids may be releted to the sterility of allotriploids. Furthermore, these results will facilitate the investigation of reproduction characteristics in allotriploids and provide theoretical basis for the study of polyploid breeding in the future.


Subject(s)
Carps , Infertility , Animals , Carps/genetics , Carps/metabolism , Female , Inhibin-beta Subunits/analysis , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Inhibins/chemistry , Male
18.
Cryo Letters ; 42(2): 67-72, 2021.
Article in English | MEDLINE | ID: mdl-33970982

ABSTRACT

BACKGROUND: Cryopreservation can induce cellular, genomic, and epigenetic abnormalities. OBJECTIVE: To analyse the impact of ovarian vitrification on follicular development and its epigenetic effect on promoter methylation of Inhba and Inhbb in granulosa cells. MATERIALS AND METHODS: Mouse ovaries were divided into control, toxicity, and vitrified groups. The growth and development of follicles were examined. After in vitro culture of follicles, DNA was extracted from isolated granulosa cells and treated with sodium bisulfite. The promoter methylation of Inhba and Inhbb was analyzed by direct PCR sequencing. RESULTS: Vitrification reduced the growth of follicles; however, antral cavity formation was not influenced negatively. Vitrification reduced the percentage of 5-methylcytosine in the Inhba promoter, while CpG sites in the promoter of Inhbb remained unmethylated. CONCLUSION: Vitrification had adverse effects on follicle growth and the epigenetics of granulosa cells. The results of the current study show that vitrification methods of ovary need more improvement.


Subject(s)
Cryopreservation , Granulosa Cells/metabolism , Inhibin-beta Subunits/metabolism , Ovarian Follicle , Vitrification , Animals , DNA Methylation , Female , Mice , Promoter Regions, Genetic
19.
Surg Today ; 51(10): 1703-1712, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33733290

ABSTRACT

PURPOSE: Papillary thyroid cancer (PTC) is generally associated with a favorable prognosis. However, some patients have fatal disease, with locally infiltrating tumors or progressive distant metastases; yet few studies have investigated the characteristics of the tumor-progressive gene expression profile in advanced PTC. We conducted this study to clarify the gene expression status in advanced PTC and identify candidate molecules for prognostic biomarkers. METHODS: We analyzed 740 tumor-progressive gene expression levels from formalin-fixed paraffin-embedded blocks of samples from six patients with low-risk PTC and six patients with high-risk PTC, using the nCounter PanCancer Progression panel. Then, we investigated the association between the expression levels of focused genes and pathological factors in PTC patients in The Cancer Genome Atlas (TCGA) database. RESULTS: The expression levels of 14 genes in the high-risk PTC specimens were more than two-fold those in the low-risk PTC specimens. In the TCGA database, expression levels of four genes (CCL11, COL6A3, INHBA, and SRPX2) were significantly higher in patients with advanced PTC. Among the patients with advanced PTC, those with high SRPX2 expression levels had poor disease-free survival. Univariate and multivariate analyses revealed that high SRPX2 expression was an independent prognostic factor. CONCLUSION: Based on the findings of this study, CCL11, COL6A3, INHBA, and SRPX2 are potential biomarkers that indicate advanced PTC. SRPX2, in particular, is considered a prognostic biomarker.


Subject(s)
Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Chemokine CCL11/genetics , Chemokine CCL11/metabolism , Collagen Type VI/genetics , Collagen Type VI/metabolism , Genetic Association Studies/methods , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Transcriptome/genetics , Adult , Aged , Disease Progression , Disease-Free Survival , Female , Gene Expression , Humans , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Male , Middle Aged , Prognosis , Risk , Thyroid Cancer, Papillary/mortality , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Young Adult
20.
Life Sci ; 284: 119222, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-33609542

ABSTRACT

OBJECTIVE: Exosomes (Exos) are known to transfer microRNAs (miRNAs) to participate in human diseases. We aim to identify the role of human umbilical cord mesenchymal stem cells (HUCMSCs)-derived Exos (HUCMSC-Exos) conveying miR-6785-5p in gastric cancer (GC). METHODS: MiR-6785-5p and inhibin subunit beta A (INHBA) expression in GC tissues and cells were determined. GC cells were transfected with the vectors that altered miR-6785-5p or INHBA expression. HUCMSCs were transfected with altered miR-6785-5p or INHBA vectors, and the HUCMSC-Exos were extracted. Then, HUCMSC-Exos were co-cultured with GC cells. The proliferation, migration, invasion, apoptosis and angiogenesis of GC cells were assessed. The binding relationship between miR-6785-5p and INHBA was verified. RESULTS: MiR-6785-5p was down-regulated and INHBA was up-regulated in GC tissues and cells. Elevation of miR-6785-5p or inhibition of INHBA restricted the malignant development of GC cells. HUCMSC-Exos suppressed malignant episodes of GC cells, which could be further enhanced by up-regulated miR-6785-5p or down-regulated INHBA. Elevated INHBA abolished the impacts of up-regulated miR-6785-5p in HUCMSC-Exos on GC cells. INHBA was confirmed as a target gene of miR-6785-5p. CONCLUSION: HUCMSC-Exos containing elevated miR-6785-5p suppress angiogenesis and metastasis in GC via inhibiting INHBA. This study may further the understanding on molecular mechanisms of GC.


Subject(s)
Exosomes/metabolism , Inhibin-beta Subunits/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , Stomach Neoplasms/blood supply , Stomach Neoplasms/genetics , Umbilical Cord/cytology , Adult , Aged , Base Sequence , Cell Proliferation/genetics , Down-Regulation/genetics , Exosomes/ultrastructure , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibin-beta Subunits/genetics , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Metastasis , Stomach Neoplasms/pathology , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL