Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
1.
Reprod Biol Endocrinol ; 20(1): 22, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35101033

ABSTRACT

BACKGROUND: Tightly regulation of extravillous cytotrophoblast (EVT) cell invasion is critical for the placentation and establishment of a successful pregnancy. Insufficient EVT cell invasion leads to the development of preeclampsia (PE) which is a leading cause of maternal and perinatal mortality and morbidity. Transforming growth factor-beta1 (TGF-ß1) and kisspeptin are expressed in the human placenta and have been shown to inhibit EVT cell invasion. Kisspeptin is a downstream target of TGF-ß1 in human breast cancer cells. However, whether kisspeptin is regulated by TGF-ß1 and mediates TGF-ß1-suppressed human EVT cell invasion remains unclear. METHODS: The effect of TGF-ß1 on kisspeptin expression and the underlying mechanisms were explored by a series of in vitro experiments in a human EVT cell line, HTR-8/SVneo, and primary cultures of human EVT cells. Serum levels of TGF-ß1 and kisspeptin in patients with or without PE were measured by ELISA. RESULTS: TGF-ß1 upregulates kisspeptin expression in HTR-8/SVneo cells and primary cultures of human EVT cells. Using pharmacological inhibitor and siRNA, we demonstrate that the stimulatory effect of TGF-ß1 on kisspeptin expression is mediated via the ALK5 receptor. Treatment with TGF-ß1 activates SMAD2/3 canonical pathways as well as ERK1/2 and PI3K/AKT non-canonical pathways. However, only inhibition of ERK1/2 activation attenuates the stimulatory effect of TGF-ß1 on kisspeptin expression. In addition, siRNA-mediated knockdown of kisspeptin attenuated TGF-ß1-suppressed EVT cell invasion. Moreover, we report that serum levels of TGF-ß1 and kisspeptin are significantly upregulated in patients with PE. CONCLUSIONS: By illustrating the potential physiological role of TGF-ß1 in the regulation of kisspeptin expression, our results may serve to improve current strategies used to treat placental diseases.


Subject(s)
Kisspeptins/genetics , Transforming Growth Factor beta1/physiology , Trophoblasts/physiology , Cell Movement/genetics , Cells, Cultured , Female , Humans , Kisspeptins/metabolism , MAP Kinase Signaling System/physiology , Pregnancy , Signal Transduction/genetics , Smad Proteins/physiology
2.
Int Urol Nephrol ; 54(3): 499-508, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35050457

ABSTRACT

OBJECTIVE: The present study aimed to investigate the effects of an imbalance in the estrogen/androgen ratio on prostate fibrosis. METHODS: Different concentrations of dihydrotestosterone (DHT) or estradiol (E2) dissolved in corn oil were injected subcutaneously into the nape of the castrated Sprague-Dawley (SD) rats over 28 consecutive days. Masson's trichrome staining and immunohistochemical staining were performed to detect the content of collagen fibers and the expression of collagen I, fibronectin, and elastin in the rat prostate of each group, respectively. DHT + E2 at different concentrations was administered to human normal prostate stromal immortalized cells (WPMY-1 cells) for 1 week. The expression of collagen I, fibronectin, elastin, transforming growth factor-ß1 (TGF-ß1), Smad3, and Smad7 was detected by Western blotting (WB). Then, WPMY-1 cells treated with 10 nM DHT + 5 pM E2 were incubated with the TGF-ß/Smad pathway inhibitor SD208 for 1 week, after which collagen I, fibronectin, and elastin expression was detected by WB. RESULTS: Compared with the uncastrated control and corn oil injection groups, the collagen fiber content and collagen I and fibronectin expression were increased and elastin expression was decreased in the castrated rat prostate with corn oil injection group (p < 0.01). Compared to castrated corn oil injection group, collagen fiber content, collagen I, and fibronectin expression were significantly decreased, and elastin expression was significantly increased in the castrated rat prostate 0.15 mg/kg DHT treatment group (p < 0.01). Following treatment with 0.15 mg/kg DHT, the content of collagen fibers, and the expression of collagen I and fibronectin were increased, and the expression of elastin was decreased in the rat prostate with increasing concentrations of E2 treatment group compared to the 0.15 mg/kg DHT group (p < 0.05, p < 0.01). Following treatment with 0.05 mg/kg E2, the collagen fiber content and the expression of collagen I and fibronectin were decreased, and the expression of elastin was increased in the rat prostate with increasing DHT concentration treatment group compared to the 0.05 mg/kg E2 group (p < 0.05, p < 0.01). Compared with the Control group, the expression of collagen I, fibronectin, TGF-ß1 and Smad3 was decreased, and the expression of elastin and Smad7 was increased in WPMY-1 cells after treatment with 10 nM DHT (p < 0.01). Following treatment with 10 nM DHT, the expression of collagen I, fibronectin, TGF-ß1, and Smad3 was increased, and the expression of elastin and Smad7 was decreased in WPMY-1 cells with increasing E2 concentration treatment compared to the 10 nM DHT group (p < 0.05, p < 0.01). Following treatment with 5 pM E2, the expression of collagen I, fibronectin, TGF-ß1, and Smad3 was decreased, and elastin and Smad7 expression was increased with increasing DHT concentration compared to the 5 pM E2 group (p < 0.05, p < 0.01). Compared to the 10 nM DHT + 5 pM E2 group, the expressions of collagen I and fibronectin were decreased; the expression of elastin was increased in WPMY-1 cells after the supplement of TGF-ß/Smad pathway inhibitor SD208 group (p < 0.05, p < 0.01). CONCLUSIONS: An imbalance in the estrogen/androgen ratio may affect prostate fibrosis. E2 may activate the degree of prostate fibrosis. In contrast to the effect of E2, DHT may inhibit the degree of prostate fibrosis, which might involve the TGF-ß/Smad signaling pathway.


Subject(s)
Androgens/analysis , Estrogens/analysis , Prostate/chemistry , Prostate/pathology , Signal Transduction/physiology , Smad Proteins/physiology , Animals , Fibrosis/etiology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta
3.
Front Immunol ; 12: 761450, 2021.
Article in English | MEDLINE | ID: mdl-34868004

ABSTRACT

IgA is the predominant antibody isotype at intestinal mucosae, where it plays a critical role in homeostasis and provides a first line of immune protection. Dysregulation of IgA production, however, can contribute to immunopathology, particularly in kidneys in which IgA deposition can cause nephropathy. Class-switch DNA recombination (CSR) to IgA is directed by TGF-ß signaling, which activates Smad2 and Smad3. Activated Smad2/Smad3 dimers are recruited together with Smad4 to the IgH α locus Iα promoter to activate germline Iα-Cα transcription, the first step in the unfolding of CSR to IgA. Epigenetic factors, such as non-coding RNAs, particularly microRNAs, have been shown to regulate T cells, dendritic cells and other immune elements, as well as modulate the antibody response, including CSR, in a B cell-intrinsic fashion. Here we showed that the most abundant miRNA in resting B cells, miR-146a targets Smad2, Smad3 and Smad4 mRNA 3'UTRs and keeps CSR to IgA in check in resting B cells. Indeed, enforced miR-146a expression in B cells aborted induction of IgA CSR by decreasing Smad levels. By contrast, upon induction of CSR to IgA, as directed by TGF-ß, B cells downregulated miR-146a, thereby reversing the silencing of Smad2, Smad3 and Smad4, which, once expressed, led to recruitment of Smad2, Smad3 and Smad4 to the Iα promoter for activation of germline Iα-Cα transcription. Deletion of miR-146a in miR-146a-/- mice significantly increased circulating levels of steady state total IgA, but not IgM, IgG or IgE, and heightened the specific IgA antibody response to OVA. In miR-146a-/- mice, the elevated systemic IgA levels were associated with increased IgA+ B cells in intestinal mucosae, increased amounts of fecal free and bacteria-bound IgA as well as kidney IgA deposition, a hallmark of IgA nephropathy. Increased germline Iα-Cα transcription and CSR to IgA in miR-146a-/- B cells in vitro proved that miR-146a-induced Smad2, Smad3 and Smad4 repression is B cell intrinsic. The B cell-intrinsic role of miR-146a in the modulation of CSR to IgA was formally confirmed in vivo by construction and OVA immunization of mixed bone marrow µMT/miR-146a-/- chimeric mice. Thus, by inhibiting Smad2, Smad3 and Smad4 expression, miR-146a plays an important and B cell intrinsic role in modulation of CSR to IgA and the IgA antibody response.


Subject(s)
Epigenesis, Genetic , Immunoglobulin A/genetics , Immunoglobulin Class Switching/genetics , MicroRNAs/physiology , Recombination, Genetic , Smad Proteins/physiology , Animals , Down-Regulation , Gastrointestinal Microbiome , Immunoglobulin A/blood , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Smad2 Protein/physiology , Smad3 Protein/physiology , Smad4 Protein/physiology
4.
Int J Oncol ; 59(4)2021 10.
Article in English | MEDLINE | ID: mdl-34476496

ABSTRACT

Epithelial membrane protein 3 (EMP3) is a transmembrane glycoprotein that contains a peripheral myelin protein 22 domain. EMP3 first received attention as a tumor suppressor, but accumulating evidence has since suggested that it may exhibit a tumor­promoting function. Nonetheless, the biological function of EMP3 remains largely unclear with regards to its role in cancer. Herein, it was shown that EMP3 expression is upregulated in non­small cell lung cancer (NSCLC) cells overexpressing aldehyde dehydrogenase 1 (ALDH1). EMP3 was shown to be involved in cell proliferation, the formation of cancer stem cells (CSCs) and in epithelial­mesenchymal transition (EMT). The ability to resist irradiation, one of the characteristics of CSCs, decreased when the EMP3 mRNA expression was knocked down using small interfering RNA. In addition, when EMP3 knockdown reduced the migratory ability of cells, a characteristic of EMT. Additionally, it was shown that the TGF­ß/Smad signaling axis was a target of EMP3. EMP3 was found to interact with TGF­ß receptor type 2 (TGFBR2) upon TGF­ß stimulation in lung CSCs (LCSC). As a result, binding of EMP3­TGFBR2 regulates TGF­ß/Smad signaling activation and consequently affects CSCs and EMT. Kaplan­Meier analysis results confirmed that patients with high expression of EMP3 had poor survival rates. Taken together, these findings showed that EMP3 may be a potential target for management of LCSCs with high expression of ALDH1, and that EMP3 is involved in TGF­ß/Smad signaling activation where it promotes acquisition of cancerous properties in tumors.


Subject(s)
Lung Neoplasms/pathology , Membrane Glycoproteins/physiology , Neoplastic Stem Cells/physiology , Transforming Growth Factor beta/physiology , Aldehyde Dehydrogenase 1 Family/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Receptor, Transforming Growth Factor-beta Type II/physiology , Signal Transduction/physiology , Smad Proteins/physiology
5.
J Zhejiang Univ Sci B ; 22(5): 410-420, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33973422

ABSTRACT

Hypertension is a prevalent systemic disease in the elderly, who can suffer from several pathological skeletal conditions simultaneously, including osteoporosis. Benidipine (BD), which is widely used to treat hypertension, has been proved to have a beneficial effect on bone metabolism. In order to confirm the osteogenic effects of BD, we investigated its osteogenic function using mouse MC3T3-E1 preosteoblast cells in vitro. The proliferative ability of MC3T3-E1 cells was significantly associated with the concentration of BD, as measured by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay and cell cycle assay. With BD treatment, the osteogenic differentiation and maturation of MC3T3-E1 cells were increased, as established by the alkaline phosphatase (ALP) activity test, matrix mineralized nodules formation, osteogenic genetic test, and protein expression analyses. Moreover, our data showed that the BMP2/Smad pathway could be the partial mechanism for the promotion of osteogenesis by BD, while BD might suppress the possible function of osteoclasts through the OPG/RANKL/RANK (receptor activator of nuclear factor-κB (NF-κB)) pathway. The hypothesis that BD bears a considerable potential in further research on its dual therapeutic effect on hypertensive patients with poor skeletal conditions was proved within the limitations of the present study.


Subject(s)
Antihypertensive Agents/pharmacology , Dihydropyridines/pharmacology , Osteogenesis/drug effects , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Protein 2/physiology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Mice , Osteoblasts/drug effects , Smad Proteins/physiology , Stem Cells/drug effects
6.
Theranostics ; 11(13): 6573-6591, 2021.
Article in English | MEDLINE | ID: mdl-33995677

ABSTRACT

Mesenchymal stem cells (MSCs) have been identified in many adult tissues. MSCs can regenerate through cell division or differentiate into adipocytes, osteoblasts and chondrocytes. As a result, MSCs have become an important source of cells in tissue engineering and regenerative medicine for bone tissue and cartilage. Several epigenetic factors are believed to play a role in MSCs differentiation. Among these, microRNA (miRNA) regulation is involved in the fine modulation of gene expression during osteogenic/chondrogenic differentiation. It has been reported that miRNAs are involved in bone homeostasis by modulating osteoblast gene expression. In addition, countless evidence has demonstrated that miRNAs dysregulation is involved in the development of osteoporosis and bone fractures. The deregulation of miRNAs expression has also been associated with several malignancies including bone cancer. In this context, bone-associated circulating miRNAs may be useful biomarkers for determining the predisposition, onset and development of osteoporosis, as well as in clinical applications to improve the diagnosis, follow-up and treatment of cancer and metastases. Overall, this review will provide an overview of how miRNAs activities participate in osteogenic/chondrogenic differentiation, while addressing the role of miRNA regulatory effects on target genes. Finally, the role of miRNAs in pathologies and therapies will be presented.


Subject(s)
Bone Diseases/genetics , Chondrogenesis/genetics , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Osteogenesis/genetics , Bone Morphogenetic Proteins/physiology , Core Binding Factor Alpha 1 Subunit/physiology , Drug Delivery Systems , Fractures, Bone/metabolism , Histone Deacetylases/physiology , Humans , Matrix Metalloproteinase 13/physiology , Repressor Proteins/physiology , Signal Transduction , Smad Proteins/physiology , Sp7 Transcription Factor/physiology , Transforming Growth Factor beta/physiology , Vascular Endothelial Growth Factor A/physiology
7.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33824966

ABSTRACT

FSH is critical for fertility. Transcription of FSHB, the gene encoding the beta subunit, is rate-limiting in FSH production and is regulated by both GnRH and activin. Activin signals through SMAD transcription factors. Although the mechanisms and importance of activin signaling in mouse Fshb transcription are well-established, activin regulation of human FSHB is less well understood. We previously reported a novel enhancer of FSHB that contains a fertility-associated single nucleotide polymorphism (rs10031006) and requires a region resembling a full (8 base-pair) SMAD binding element (SBE). Here, we investigated the role of the putative SBE within the enhancer in activin and GnRH regulation of FSHB. In mouse gonadotrope-derived LßT2 cells, the upstream enhancer potentiated activin induction of both the human and mouse FSHB proximal promoters and conferred activin responsiveness to a minimal promoter. Activin induction of the enhancer required the SBE and was blocked by the inhibitory SMAD7, confirming involvement of the classical SMAD signaling pathway. GnRH induction of FSHB was also potentiated by the enhancer and dependent on the SBE, consistent with known activin/GnRH synergy regulating FSHB transcription. In DNA pull-down, the enhancer SBE bound SMAD4, and chromatin immunoprecipitation demonstrated SMAD4 enrichment at the enhancer in native chromatin. Combined activin/GnRH treatment elevated levels of the active transcriptional histone marker, histone 3 lysine 27 acetylation, at the enhancer. Overall, this study indicates that the enhancer is directly targeted by activin signaling and identifies a novel, evolutionarily conserved mechanism by which activin and GnRH can regulate FSHB transcription.


Subject(s)
Activins/pharmacology , Enhancer Elements, Genetic/physiology , Follicle Stimulating Hormone, beta Subunit/genetics , Gonadotropin-Releasing Hormone/pharmacology , Transcription, Genetic/drug effects , Activins/metabolism , Animals , Drug Synergism , Enhancer Elements, Genetic/drug effects , Follistatin/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Humans , Mice , Promoter Regions, Genetic/drug effects , Signal Transduction , Smad Proteins/physiology , Smad4 Protein/metabolism
8.
Clin Exp Metastasis ; 38(2): 139-161, 2021 04.
Article in English | MEDLINE | ID: mdl-33590419

ABSTRACT

Epithelial ovarian cancer (EOC) is a leading cause of cancer-related death in women. Late-stage diagnosis with significant tumor burden, accompanied by recurrence and chemotherapy resistance, contributes to this poor prognosis. These morbidities are known to be tied to events associated with epithelial-mesenchymal transition (EMT) in cancer. During EMT, localized tumor cells alter their polarity, cell-cell junctions, cell-matrix interactions, acquire motility and invasiveness and an exaggerated potential for metastatic spread. Key triggers for EMT include the Transforming Growth Factor-ß (TGFß) family of growth factors which are actively produced by a wide array of cell types within a specific tumor and metastatic environment. Although TGFß can act as either a tumor suppressor or promoter in cancer, TGFß exhibits its pro-tumorigenic functions at least in part via EMT. TGFß regulates EMT both at the transcriptional and post-transcriptional levels as outlined here. Despite recent advances in TGFß based therapeutics, limited progress has been seen for ovarian cancers that are in much need of new therapeutic strategies. Here, we summarize and discuss several recent insights into the underlying signaling mechanisms of the TGFß isoforms in EMT in the unique metastatic environment of EOCs and the current therapeutic interventions that may be relevant.


Subject(s)
Carcinoma, Ovarian Epithelial/pathology , Ovarian Neoplasms/pathology , Transforming Growth Factor beta/physiology , Carcinoma, Ovarian Epithelial/drug therapy , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Neoplasm Metastasis , Ovarian Neoplasms/drug therapy , Signal Transduction/physiology , Smad Proteins/physiology , Transforming Growth Factor beta/antagonists & inhibitors
10.
Pediatr Res ; 89(3): 502-509, 2021 02.
Article in English | MEDLINE | ID: mdl-32365352

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia remains one of the most common complications of prematurity, despite significant improvements in perinatal care. Functional modeling of human lung development and disease, like BPD, is limited by our ability to access the lung and to maintain relevant progenitor cell populations in culture. METHODS: We supplemented Rho/SMAD signaling inhibition with mTOR inhibition to generate epithelial basal cell-like cell lines from tracheal aspirates of neonates. RESULTS: Single-cell RNA-sequencing confirmed the presence of epithelial cells in tracheal aspirates obtained from intubated neonates. Using Rho/SMAD/mTOR triple signaling inhibition, neonatal tracheal aspirate-derived (nTAD) basal cell-like cells can be expanded long term and retain the ability to differentiate into pseudostratified airway epithelium. CONCLUSIONS: Our data demonstrate that neonatal tracheal aspirate-derived epithelial cells can provide a novel ex vivo human cellular model to study neonatal lung development and disease. IMPACT: Airway epithelial basal cell-like cell lines were derived from human neonatal tracheal aspirates. mTOR inhibition significantly extends in vitro proliferation of neonatal tracheal aspirate-derived basal cell-like cells (nTAD BCCs). nTAD BCCs can be differentiated into functional airway epithelium. nTAD BCCs provide a novel model to investigate perinatal lung development and diseases.


Subject(s)
Epithelial Cells/drug effects , Smad Proteins/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Trachea/cytology , rho-Associated Kinases/antagonists & inhibitors , Base Sequence , Body Fluids/cytology , Bronchopulmonary Dysplasia , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Epithelial Cells/chemistry , Epithelial Cells/cytology , Humans , Infant, Newborn , Primary Cell Culture , Single-Cell Analysis , Sirolimus/pharmacology , Smad Proteins/physiology , Stem Cells/cytology , Stem Cells/drug effects , Suction , TOR Serine-Threonine Kinases/physiology , rho-Associated Kinases/physiology
11.
J Orthop Res ; 39(6): 1297-1304, 2021 06.
Article in English | MEDLINE | ID: mdl-32043642

ABSTRACT

Heterotopic ossification (HO) is a pathological condition in which ectopic bone forms within soft tissues such as skeletal muscle. Human platelet-derived growth factor receptor α positive (PDGFRα+) cells, which were proved to be the original cells of HO were incubated in osteogenic differentiation medium with Food and Drug Administration-approved compounds. Alkaline phosphatase activity was measured as a screening to inhibit osteogenic differentiation. For the compounds which inhibited osteogenic differentiation of PDGFRα+ cells, we examined dose dependency of its effect using alizarin red S staining and its cell toxicity using WST-8. In addition, regulation of bone morphogenetic proteins (BMP)-Smad signaling which is the major signal of osteogenic differentiation was investigated by Western blotting to elucidate the mechanism of osteogenesis inhibitory effect by the compound. In vivo experiment, complete transverse incision of Achilles tendons in mice was made and mice were fed the compound by mixing with drinking water after operation. Ten weeks after operation, we assessed and quantified HO by micro-computed tomography scan. Intriguingly, we discovered desloratadine inhibited osteogenic differentiation of PDGFRα+ cells using the drug repositioning method. Desloratadine inhibited osteogenic differentiation of the cells dose dependently without cell toxicity. Desloratadine suppressed phosphorylation of Smad1/5/8 induced by BMP2 in PDGFRα+ cells. In Achilles tenotomy mice model, desloratadine treatment significantly inhibited ectopic bone formation compared with control. In conclusion, we discovered desloratadine inhibited osteogenic differentiation using human PDGFRα+ cells and proved its efficacy using Achilles tenotomy ectopic bone formation model in vivo. Our study paved the way to inhibit HO in early clinical use because of its guaranteed safety.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Loratadine/analogs & derivatives , Ossification, Heterotopic/prevention & control , Smad Proteins/physiology , Animals , Cell Differentiation/drug effects , Loratadine/pharmacology , Loratadine/therapeutic use , Male , Mice , Mice, Inbred C57BL , Osteogenesis/drug effects , Receptor, Platelet-Derived Growth Factor alpha/analysis , Signal Transduction/drug effects
12.
Pharm Biol ; 58(1): 1244-1251, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33332205

ABSTRACT

CONTEXT: Oral submucous fibrosis (OSF) is a chronic and progressive disease. Arecoline, present in betel nuts, has been proposed as a vital aetiological factor. However, the underlying mechanism remains unclear. OBJECTIVES: This research elucidates the expression of tropomyosin-1 (TPM1) and its regulation mechanism in HaCaT cells treated with arecoline. MATERIALS AND METHODS: HaCaT cells were assigned into three groups: (1) Control; (2) Treated with arecoline (0.16 mM) for 48 h (3) Treated with arecoline (0.16 mM) and transfected with small interfering RNA (siRNA) for TPM1 (50 nM) for 48 h. CCK8, cell cycle, and apoptosis phenotypic analyses were performed. PCR and western blot analyses were performed to detect the expression level of TPM1 and examine the related signalling pathway. RESULTS: The IC50 of arecoline was approximately 50 µg/mL (0.21 mM). The arecoline dose (0.16 mM) and time (48 h) markedly increased TPM1 expression at the mRNA and protein levels in HaCaT cells. Arecoline suppressed the cell growth, caused cell cycle arrest at the G1 phase, and induced cell apoptosis in HaCaT cells. siRNA-mediated knockdown of TPM1 attenuated the effect of arecoline on cell proliferation, apoptosis, and cell cycle arrest at the G1 phase. Furthermore, blocking of the transforming growth factor (TGF)-ß receptor using SB431542 significantly suppressed TPM1 expression in the cells treated with arecoline. DISCUSSION AND CONCLUSIONS: Arecoline suppresses HaCaT cell viability by upregulating TPM1 through the TGF-ß/Smad signalling pathway. This research provides a scientific basis for further study of arecoline and TPM1 in OSF and can be generalised to broader pharmacological studies. TPM1 may be a promising molecular target for treating OSF.


Subject(s)
Arecoline/toxicity , Oral Submucous Fibrosis/chemically induced , Smad Proteins/physiology , Transforming Growth Factor beta/physiology , Tropomyosin/genetics , Apoptosis/drug effects , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/physiology , HaCaT Cells , Humans , Signal Transduction/drug effects , Signal Transduction/physiology , Tropomyosin/physiology , Up-Regulation
13.
Biomed Pharmacother ; 131: 110746, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33152915

ABSTRACT

This study investigated the therapeutic efficacy of Bu-Shen-Jiang-Ya decoction (BSJYD) on hypertensive renal damage to determine whether it regulates the expression of transforming growth factor-ß (TGF-ß)/SMADs signaling pathways, thereby relieving renal fibrosis in Dahl salt-sensitive (SS) rats. Dahl SS rats on a high-sodium diet were prospectively treated with BSJYD (n = 12) or valsartan (n = 12) for 8 weeks. The blood pressure (BP) of these rats was measured and their kidneys were subjected to biochemical analysis, including serum creatinine (Scr) and blood urea nitrogen (BUN); hematoxylin and eosin staining; Masson trichrome staining; real-time polymerase chain reaction; and western blot analysis. The primary outcome was that BSJYD significantly reduced BP, debased BUN, and Scr and ameliorated renal pathological changes. As underlying therapeutic mechanisms, BSJYD reduces TGFß1 and Smad2/3 expression and suppresses renal fibrosis, as suggested by the decreased expression of connective tissue growth factor(CTGF). These data suggest that BSJYD acts as an optimal therapeutic agent for hypertensive renal damage by inhibiting the TGF-ß/SMADs signaling pathway.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Hypertension/drug therapy , Kidney Diseases/prevention & control , Medicine, Chinese Traditional , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Blood Pressure/drug effects , Connective Tissue Growth Factor/genetics , Drugs, Chinese Herbal/pharmacology , Hypertension/complications , Hypertension/pathology , Kidney/pathology , Male , Rats , Rats, Inbred Dahl , Signal Transduction/drug effects , Smad Proteins/genetics , Smad Proteins/physiology , Transforming Growth Factor beta/physiology
14.
Biomed Pharmacother ; 127: 110132, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32403042

ABSTRACT

BACKGROUND: Myocardial fibrosis is an important pathological feature of pressure overload cardiac remodeling. Si-Miao-Yong-An decoction (SMYAD), a traditional Chinese formula, is now clinically used in the treatment of cardiovascular diseases in China. However, its mechanisms in the prevention of heart failure are not fully revealed. PURPOSE: To determine whether treatment with SMYAD for 4 weeks would lead to changes in collagen metabolism and ventricular remodeling in a mice model of heart failure. METHODS: Mice were subjected to transverse aorta constriction to generate pressure overload induced cardiac remodeling and then were administered SMYAD (14.85 g/kg/day) or captopril (16.5 mg/kg/day) intragastrically for 4 weeks after surgery. Echocardiography and immunohistochemical examination were used to evaluate the effects of SMYAD. The mRNA of collagen metabolism biomarkers were detected. Protein expression of TGF-ß1/Smad and TGF-ß1/TAK1/p38 pathway were assessed by Western blot. RESULTS: SMYAD significantly improved cardiac function, increased left ventricle ejection fraction, and decreased fibrosis area and αSMA expression. Moreover, SMYAD reduced proteins expression related to collagen metabolism, including Col1, Col3, TIMP2 and CTGF. The increased levels of TGF-ß1, Smad2, and Smad3 phosphorylation were attenuated in SMYAD group. In addition, SMYAD reduced the levels of TGF-ß1, p-TAK1 and p-p38 compared with TAC group. CONCLUSIONS: SMYAD improved cardiac fibrosis and heart failure by inhibition of TGF-ß1/Smad and TGF-ß1/TAK1/p38 pathway. SMYAD protected against cardiac fibrosis and maintained collagen metabolism balance by regulating MMP-TIMP expression. Taken together, these results indicate that SMYAD might be a promising therapeutic agent against cardiac fibrosis.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Heart Failure/drug therapy , Matrix Metalloproteinases/metabolism , Myocardium/pathology , Tissue Inhibitor of Metalloproteinases/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Collagen/metabolism , Drugs, Chinese Herbal/therapeutic use , Fibrosis , MAP Kinase Kinase Kinases/physiology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Smad Proteins/physiology , Transforming Growth Factor beta1/physiology , Ventricular Remodeling/drug effects
15.
Oncogene ; 39(22): 4436-4449, 2020 05.
Article in English | MEDLINE | ID: mdl-32350443

ABSTRACT

Activator protein (AP)-1 transcription factors are essential elements of the pro-oncogenic functions of transforming growth factor-ß (TGFß)-SMAD signaling. Here we show that in multiple HER2+ and/or EGFR+ breast cancer cell lines these AP-1-dependent tumorigenic properties of TGFß critically rely on epidermal growth factor receptor (EGFR) activation and expression of the ΔN isoform of transcriptional regulator p63. EGFR and ΔNp63 enabled and/or potentiated the activation of a subset of TGFß-inducible invasion/migration-associated genes, e.g., ITGA2, LAMB3, and WNT7A/B, and enhanced the recruitment of SMAD2/3 to these genes. The TGFß- and EGF-induced binding of SMAD2/3 and JUNB to these gene loci was accompanied by p63-SMAD2/3 and p63-JUNB complex formation. p63 and EGFR were also found to strongly potentiate TGFß induction of AP-1 proteins and, in particular, FOS family members. Ectopic overexpression of FOS could counteract the decrease in TGFß-induced gene activation after p63 depletion. p63 is also involved in the transcriptional regulation of heparin binding (HB)-EGF and EGFR genes, thereby establishing a self-amplification loop that facilitates and empowers the pro-invasive functions of TGFß. These cooperative pro-oncogenic functions of EGFR, AP-1, p63, and TGFß were efficiently inhibited by clinically relevant chemical inhibitors. Our findings may, therefore, be of importance for therapy of patients with breast cancers with an activated EGFR-RAS-RAF pathway.


Subject(s)
Breast Neoplasms/pathology , Epidermal Growth Factor/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Neoplasm Proteins/physiology , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Transcription, Genetic , Transforming Growth Factor beta1/physiology , Tumor Suppressor Proteins/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , ErbB Receptors/physiology , Female , Humans , MAP Kinase Signaling System , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-fos/physiology , Proto-Oncogene Proteins c-jun/physiology , Receptor, ErbB-2/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Smad Proteins/physiology
16.
Pancreas ; 49(4): 534-542, 2020 04.
Article in English | MEDLINE | ID: mdl-32282767

ABSTRACT

OBJECTIVES: Transforming growth factor ß (TGF-ß) signaling pathway is one of the core pathways in pancreatic ductal adenocarcinoma (PDAC). Prognostic value of TGF-ß pathway genes as a functionally related group in PDAC is rarely studied. METHODS: Seventy-two PDAC patients who underwent surgery between November 30, 2015, and September 13, 2017, in West China Hospital, Sichuan University, were identified and included in this study. Whole-exome sequencing or targeted next-generation sequencing was performed with tumor tissue. Clinicopathologic characteristics and survival data were retrospectively collected and analyzed. RESULTS: Genetic alterations were detected in 71 patients (98.6%). Although 1 patient (1.4%) had one genetic alteration, 33 patients (45.8%) had 2 to 4 alterations and 37 patients (51.4%) had 5 or more alterations. Twenty-five patients with TGF-ß pathway alteration were identified as TGF-ßm+ group. Other 47 patients were TGF-ßm- group. Mutation of TGF-ß pathway was independently associated with inferior survival (hazard ratio, 2.22, 95% confidence interval, 1.05-4.70, P = 0.04), especially in patients accepting radical surgery (hazard ratio, 3.25, 95% confidence interval, 1.01-10.49, P = 0.04). CONCLUSIONS: Inferior prognosis was observed in PDACs with mutations of TGF-ß pathway. Genomic information could help screen out patients at risk after surgery, and adjuvant therapy might benefit this subgroup of PDACs.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Neoplasm Proteins/physiology , Pancreatic Neoplasms/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Adult , Aged , Asian People/genetics , Brachytherapy , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/surgery , DNA, Neoplasm/genetics , Female , Genes, Neoplasm , High-Throughput Nucleotide Sequencing , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Proteins/genetics , Palliative Care , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Prognosis , Proportional Hazards Models , Radiotherapy, Adjuvant , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/physiology , Retrospective Studies , Smad Proteins/genetics , Smad Proteins/physiology , Transforming Growth Factor beta/genetics , Exome Sequencing
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 51(2): 165-170, 2020 Mar.
Article in Chinese | MEDLINE | ID: mdl-32220183

ABSTRACT

OBJECTIVE: To investigate the regulation of fibromodulin (FMOD) on proliferation, adhesion and migration of non-small cell lung cancer cell line H322, and discuss its action mechanism. METHODS: H322 cells were randomly divided into control group, small interfering RNA (siRNA) silencing FMOD ( FMOD siRNA) group and control siRNA (Con siRNA) group. FMOD siRNA and Con siRNA were transfected into H322 cells. The cell viability of each group was detected by CCK-8 method. The adhesion ability of cells was detected by fluorescein diacetate (FDA) fluorescent staining. The cell migration ability was detected by Transwell method. Real time-PCR was used to detect the mRNA expressions of Cyclin D1, intercellular adhesion molecule -1 (ICAM-1), E-cadherin, FMOD, transforming growth factor-ß (TGF-ß), Smad2, Smad3, Smad4 and Smad7 in cells. The protein expressions of Cyclin D1, ICAM-1, E-cadherin, FMOD, TGF-ß1, Smad2, Smad3, Smad4 and Smad7 were detected by Western blot. RESULTS: Compared with the Con siRNA group, the cell viability, cell adhesion and migration ability of the FMOD siRNA group were decreased, and the difference was statistically significant ( P<0.01). There was no significant difference between the control group and the Con siRNA group. Real time-PCR and Western blot results showed that the mRNA and protein expression levels of Cyclin D1, ICAM-1, TGF-ß1, Smad2, Smad3 and Smad4 were decreased in FMOD siRNA group, compared with Con siRNA group, while the mRNA and protein expression levels of E-cadherin and Smad7 are elevated. CONCLUSION: Silencing of the FMOD gene significantly reduces the proliferation, adhesion and migration of H322 cells, which may be conducted by inhibiting the TGF-ß/Smad signaling pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Fibromodulin/genetics , Gene Silencing , Lung Neoplasms , Smad Proteins , Transforming Growth Factor beta , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Adhesion , Cell Movement , Cell Proliferation , Fibromodulin/physiology , Gene Expression , Humans , Lung Neoplasms/metabolism , RNA, Small Interfering , Signal Transduction , Smad Proteins/physiology , Transforming Growth Factor beta/physiology
18.
Invest Ophthalmol Vis Sci ; 61(2): 1, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32031573

ABSTRACT

Purpose: This study aimed to explore the role of the protein kinase A (PKA) pathway in proliferative vitreoretinopathy (PVR) and the effect of the PKA inhibitor H89 on experimental PVR. Methods: Epiretinal membranes (ERMs) were acquired from PVR patients and analyzed by frozen-section immunofluorescence. An in vivo model was developed by intravitreal injecting rat eyes with ARPE-19 cells and platelet-rich plasma, and changes in eye structures and vision function were observed. An in vitro epithelial-mesenchymal transition (EMT) cell model was established by stimulating ARPE-19 cells with transforming growth factor (TGF)-ß. Alterations in EMT-related genes and cell function were detected. Mechanistically, PKA activation and activity were explored to assess the relationship between TGF-ß1 stimulation and the PKA pathway. The effect of H89 on the TGF-ß-Smad2/3 pathway was detected. RNA sequencing was used to analyze gene expression profile changes after H89 treatment. Results: PKA was activated in human PVR membranes. In vivo, H89 treatment protected against structural changes in the retina and prevented decreases in electroretinogram b-wave amplitudes. In vitro, H89 treatment inhibited EMT-related gene alterations and partially reversed the functions of the cells. TGF-ß-induced PKA activation was blocked by H89 pretreatment. H89 did not affect the phosphorylation or nuclear translocation of regulatory Smad2/3 but increased the expression of inhibitory Smad6. Conclusions: PKA pathway activation is involved in PVR pathogenesis, and the PKA inhibitor H89 can effectively inhibit PVR, both in vivo and in vitro. Furthermore, the protective effect of H89 is related to an increase in inhibitory Smad6.


Subject(s)
Isoquinolines/antagonists & inhibitors , Sulfonamides/antagonists & inhibitors , Vitreoretinopathy, Proliferative/drug therapy , Aged , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Electroretinography , Epiretinal Membrane/metabolism , Epithelial Cells/metabolism , Female , Humans , Isoquinolines/pharmacology , MAP Kinase Signaling System/physiology , Male , Middle Aged , Retinal Pigment Epithelium/drug effects , Smad Proteins/physiology , Sulfonamides/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors
19.
Acta Trop ; 205: 105307, 2020 May.
Article in English | MEDLINE | ID: mdl-31862462

ABSTRACT

Excretory/Secretory products (ESPs) from Clonorchis sinensis-a fluke dwelling on the biliary ducts-promote the activation of hepatic stellate cells (HSCs) and lead to hepatic fibrosis ultimately, although the mechanisms that are responsible for CsESPs-induced activation of HSCs are largely unknown. In the present study, we investigated the underlying mechanism of TLR4 in the regulation of the activation of HSCs caused by CsESPs. We found that the expression of TLR4 was significantly increased in the HSCs with CsESPs for 24 h, compared to the control group. However, the activation of HSCs induced by CsESPs was inhibited by interfering with TGF-ß/Smad pathway using a TGF-ß receptor I inhibitor LY2157299, indicating that TGF-ß induced signaling pathway was involved in CsESPs-caused the activation of HSCs. In addition, the activation of HSCs caused by CsESPs was remarkably inhibited by a TLR4 specific inhibitor (VIPER), and phosphorylation of Smad2/3 was significantly attenuated but the expression of the pseudoreceptor of TGF-ß-type I receptor (BAMBI) was obviously increased when TLR4 signaling pathway was blocked. The results of the present study demonstrate that activation of HSCs caused by CsESPs is mediated by a cross-talk between TLR4 and TGF-ß/Smads signaling pathway, and may provide a potential treatment strategy to interrupt the process of liver fibrosis caused by C. sinensis.


Subject(s)
Clonorchis sinensis/pathogenicity , Hepatic Stellate Cells/physiology , Liver Cirrhosis/etiology , Smad Proteins/physiology , Toll-Like Receptor 4/physiology , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Rabbits , Signal Transduction/physiology
20.
J Am Soc Nephrol ; 30(11): 2073-2090, 2019 11.
Article in English | MEDLINE | ID: mdl-31653783

ABSTRACT

BACKGROUND: Necroptosis is a newly discovered cell death pathway that plays a critical role in AKI. The involvement of integrin-linked kinase (ILK) in necroptosis has not been studied. METHODS: We performed experiments in mice with an Ilk deletion in collecting duct (CD) principal cells (PCs), and cultured tubular epithelial cells treated with an ILK inhibitor or ILK siRNA knockdown. RESULTS: Ilk deletion in CD PCs resulted in acute tubular injury and early mortality in mice. Progressive interstitial fibrosis and inflammation associated with the activation of the canonical TGF-ß signaling cascade were detected in the kidneys of the mice lacking ILK in the CD PCs. In contrast to the minimal apoptosis detected in the animals' injured CDs, widespread necroptosis was present in ILK-deficient PCs, characterized by cell swelling, deformed mitochondria, and rupture of plasma membrane. In addition, ILK deficiency resulted in increased expression and activation of necroptotic proteins MLKL and RIPK3, and membrane translocation of MLKL in CD PCs. ILK inhibition and siRNA knockdown reduced cell survival in cultured tubular cells, concomitant with increased membrane accumulation of MLKL and/or phospho-MLKL. Administration of a necroptosis inhibitor, necrostatin-1, blocked cell death in vitro and significantly attenuated inflammation, interstitial fibrosis, and renal failure in ILK-deficient mice. CONCLUSIONS: The study demonstrates the critical involvement of ILK in necroptosis through modulation of the RIPK3 and MLKL pathway and highlights the contribution of CD PC injury to the development of inflammation and interstitial fibrosis of the kidney.


Subject(s)
Kidney Tubules, Collecting/pathology , Kidney/pathology , Necroptosis , Nephritis/etiology , Protein Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , Fibrosis , Mice , Mice, Inbred C57BL , Protein Kinases/physiology , Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Smad Proteins/physiology , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...