Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 223
Filter
1.
Cell Prolif ; 55(10): e13310, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35920128

ABSTRACT

OBJECTIVE: GDF11 is a member of the TGF-ß superfamily that was recently implicated as potential "rejuvenating" factor, which can ameliorate metabolic disorders. The main objective of the presented study was to closely characterize the role of GDF11 signaling in the glucose homeostasis and in the differentiation of white adipose tissue. METHODS: We performed microscopy imaging, biochemical and transcriptomic analyses of adipose tissues of 9 weeks old ob/ob mice and murine and human pre-adipocyte cell lines. RESULTS: Our in vivo experiments employing GDF11 treatment in ob/ob mice showed improved glucose/insulin homeostasis, decreased weight gain and white adipocyte size. Furthermore, GDF11 treatment inhibited adipogenesis in pre-adipocytes by ALK5-SMAD2/3 activation in cooperation with the WNT/ß-catenin pathway, whose inhibition resulted in adipogenic differentiation. Lastly, we observed significantly elevated levels of the adipokine hormone adiponectin and increased glucose uptake by mature adipocytes upon GDF11 exposure. CONCLUSION: We show evidence that link GDF11 to adipogenic differentiation, glucose, and insulin homeostasis, which are pointing towards potential beneficial effects of GDF11-based "anti-obesity" therapy.


Subject(s)
Adipogenesis , beta Catenin , Adipocytes/metabolism , Adiponectin/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Glucose/metabolism , Growth Differentiation Factors/metabolism , Humans , Insulin/metabolism , Mice , Receptor, Transforming Growth Factor-beta Type I , Smad Proteins, Receptor-Regulated , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
2.
Cell Rep ; 40(2): 111038, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830803

ABSTRACT

Despite the fundamental roles of TGF-ß family signaling in cell fate determination in all metazoans, the mechanism by which these signals are spatially and temporally interpreted remains elusive. The cell-context-dependent function of TGF-ß signaling largely relies on transcriptional regulation by SMAD proteins. Here, we discover that the DNA repair-related protein, HMCES, contributes to early development by maintaining nodal/activin- or BMP-signaling-regulated transcriptional network. HMCES binds with R-SMAD proteins, co-localizing at active histone marks. However, HMCES chromatin occupancy is independent on nodal/activin or BMP signaling. Mechanistically, HMCES competitively binds chromatin to limit binding by R-SMAD proteins, thereby forcing their dissociation and resulting in repression of their regulatory effects. In Xenopus laevis embryo, hmces KD causes dramatic development defects with abnormal left-right axis asymmetry along with increasing expression of lefty1. These findings reveal HMCES transcriptional regulatory function in the context of TGF-ß family signaling.


Subject(s)
Activins , Bone Morphogenetic Proteins , Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Chromatin , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
3.
Biomed Pharmacother ; 146: 112499, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34959122

ABSTRACT

Atherosclerosis (AS) is a chronic inflammatory vascular disease with a multifactorial pathogenesis. It becomes a global health concern, especially causing an array of fatal consequences among the elderly. However, the mechanisms of AS remain unexplained. The transforming growth factor-ß (TGF-ß) signaling pathway is widely involved in the inflammation, immune function, proliferation, differentiation,and apoptosis in vivo. Based on previous researches, it has not been confirmed whether the TGF-ß pathway promotes or inhibits atherosclerosis. Furthermore, more and more studies have found that microRNAs can regulate atherosclerosis through the TGF-ß signaling pathway. In this review, we summarize and discuss the role of microRNAs in the pathogenesis of atherosclerosis via the TGF-ß signaling pathway.


Subject(s)
Atherosclerosis/pathology , MicroRNAs/metabolism , Transforming Growth Factor beta/metabolism , Apoptosis/physiology , Cell Proliferation/physiology , Down-Regulation , Endothelium, Vascular/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress , Signal Transduction/physiology , Smad Proteins, Receptor-Regulated/metabolism , Up-Regulation
4.
Lab Invest ; 101(11): 1475-1483, 2021 11.
Article in English | MEDLINE | ID: mdl-34504305

ABSTRACT

Oral malignant melanoma, which frequently invades the hard palate or maxillary bone, is extremely rare and has a poor prognosis. Bone morphogenetic protein (BMP) is abundantly expressed in bone matrix and is highly expressed in malignant melanoma, inducing an aggressive phenotype. We examined the role of BMP signaling in the acquisition of an aggressive phenotype in melanoma cells in vitro and in vivo. In five cases, immunohistochemistry indicated the phosphorylation of Smad1/5 (p-Smad1/5) in the nuclei of melanoma cells. In the B16 mouse and A2058 human melanoma cell lines, BMP2, BMP4, or BMP7 induces morphological changes accompanied by the downregulation of E-cadherin, and the upregulation of N-cadherin and Snail, markers of epithelial-mesenchymal transition (EMT). BMP2 also stimulates cell invasion by increasing matrix metalloproteinase activity in B16 cells. These effects were canceled by the addition of LDN193189, a specific inhibitor of Smad1/5 signaling. In vivo, the injection of B16 cells expressing constitutively activated ALK3 enhanced zygoma destruction in comparison to empty B16 cells by increasing osteoclast numbers. These results suggest that the activation of BMP signaling induces EMT, thus driving the acquisition of an aggressive phenotype in malignant melanoma.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Bone Neoplasms/secondary , Melanoma/secondary , Mouth Neoplasms/pathology , Smad Proteins, Receptor-Regulated/metabolism , Animals , Bone Neoplasms/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Humans , Male , Melanoma/metabolism , Mice , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Signal Transduction
5.
J Pharm Pharmacol ; 73(11): 1442-1450, 2021 Oct 07.
Article in English | MEDLINE | ID: mdl-34128987

ABSTRACT

OBJECTIVES: Hyperglycemia-induced SIRT1, DNMT1, SODs, as well as oxidative stress, play a pivotal role in the progression of diabetic nephropathy. Cissus quadrangularis, holds antioxidant and hypoglycemic activity; however, a direct link between its activity and prevention of diabetic nephropathy has not been ascertained yet. Accordingly, we aimed to delineate the protective effect of ethanolic extract of Cissus quadrangularis (EECQ) against high-fat diet/streptozotocin (HFD/STZ) induced diabetic nephropathy rats. METHODS: The control group was fed with a normal chow diet. Rats kept on an HFD for 12 weeks with a single low dose of STZ manifested the features of diabetic nephropathy. The treatment was done by the oral administration of EECQ (200 mg/kg) for six weeks (six rats in each group). KEY FINDINGS: Treatment with EECQ demonstrated substantial attenuation of elevated insulin resistance, lipid profile and creatinine level. Additionally, EECQ restored albuminuria, glomerular filtration rate and creatinine clearance in diabetic nephropathy rats. Furthermore, HFD consumption in rats culminated in reduced SIRT1 and enhanced DNMT1 expression, nonetheless, rescued by EECQ. Moreover, EECQ augmented the SOD 1 and 3 levels, thereby safeguarded from oxidative damage and renal inflammation. Besides, treatment protected from renal fibrosis by downregulating TGFß, Smad2/3 and col1/3 expression in diseased rats. CONCLUSIONS: Thus, based on the above findings, we conclude that EECQ shows a protective effect against diabetic nephropathy.


Subject(s)
Cissus , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Kidney/drug effects , Plant Extracts/pharmacology , Repressor Proteins/metabolism , Sirtuin 1/metabolism , Albuminuria , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Creatinine/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/prevention & control , Diet, High-Fat , Glomerular Filtration Rate , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Inflammation , Insulin Resistance , Kidney/metabolism , Kidney/pathology , Lipids/blood , Male , Oxidative Stress/drug effects , Phytotherapy , Plant Extracts/therapeutic use , Rats, Sprague-Dawley , Smad Proteins, Receptor-Regulated/metabolism , Superoxide Dismutase/metabolism , Transforming Growth Factor beta/metabolism
6.
Lab Invest ; 101(9): 1166-1175, 2021 09.
Article in English | MEDLINE | ID: mdl-34168289

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays a crucial role in the development of pulmonary fibrosis. This study aims to investigate the effects of valproic acid (VPA) on EMT in vitro and in vivo. In vitro, EMT was induced by the administration of transforming growth factor-ß1 (TGF-ß1) in a human alveolar epithelial cell line (A549). The dose effects of VPA (0.1-3 mM) on EMT were subsequently evaluated at different timepoints. VPA (1 mM) was applied prior to the administration of TGF-ß1 and the expression of E-cadherin, vimentin, p-Smad2/3 and p-Akt was assessed. In addition, the effects of a TGF-ß type I receptor inhibitor (A8301) and PI3K-Akt inhibitor (LY294002) on EMT were evaluated. In vivo, the effects of VPA on bleomycin-induced lung fibrosis were evaluated by assessing variables such as survival rate, body weight and histopathological changes, whilst the expression of E-cadherin and vimentin in lung tissue was also evaluated. A8301 and LY294002 were used to ascertain the cellular signaling pathways involved in this model. The administration of VPA prior to TGF-ß1 in A549 cells prevented EMT in both a time- and concentration-dependent manner. Pretreatment with VPA downregulated the expression of both p-Smad2/3 and p-Akt. A8301 administration increased the expression of E-cadherin and reduced the expression of vimentin. LY294002 inhibited Akt phosphorylation induced by TGF-ß1 but failed to prevent EMT. Pretreatment with VPA both increased the survival rate and prevented the loss of body weight in mice with pulmonary fibrosis. Interestingly, both VPA and A8301 prevented EMT and facilitated an improvement in lung structure. Overall, pretreatment with VPA attenuated the development of pulmonary fibrosis by inhibiting EMT in mice, which was associated with Smad2/3 deactivation but without Akt cellular signal involvement.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Lung/drug effects , Pulmonary Fibrosis/metabolism , Valproic Acid/pharmacology , A549 Cells , Animals , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta1/metabolism
7.
Cell Cycle ; 20(12): 1147-1162, 2021 06.
Article in English | MEDLINE | ID: mdl-34110955

ABSTRACT

Ovarian cancer (OC) is the fifth most common female malignant tumor and the leading cause of cancer-related death in women worldwide. Epithelial ovarian cancer (EOC) is the predominant type of OC. Investigating the mechanism underlying tumorigenesis and progression of EOC is urgent. Our previous research has shown that long non-coding RNAs (lncRNAs) CDKN2A-AS1 is upregulated in EOC tissues and cells. Furthermore, we have predicted that CDKN2A-AS1 is associated with the bone morphogenetic protein (BMP)-SMAD signaling pathway, which is negatively regulated by the sclerostin domain containing 1 (SOSTDC1). Therefore, we conjecture that the CDKN2A-AS1 regulate BMP-SMAD signaling pathway via interacting with SOSTDC1, which need more investigation. Moreover, the functions of the BMP-SMAD signaling pathway and the SOSTDC1 on EOC are still unclear. Herein, we unearthed that CDKN2A-AS1, BMP2/4/7, SMAD1/5/9 and phosphorylation of SMAD1/5/9 (p-SMAD1/5/9) were upregulated in EOC tissues and cells, whereas SOSTDC1 was downregulated in EOC tissues and cells. We firstly demonstrated that CDKN2A-AS1 bound directly with the SOSTDC1. CDKN2A-AS1 downregulated the expression of SOSTDC1, but upregulated the expression of BMP2/4/7, SMAD1/5/9, and p-SMAD1/5/9. CDKN2A-AS1 promoted the proliferation, migration, invasion of EOC cells and tumor growth in vivo, whereas SOSTDC1 inhibited the proliferation, migration, invasion of EOC cells. Knockdown SOSTDC1 rescued the inhibitory effect of si-lncRNA CDKN2A-AS1 on the EOC cells proliferation, migration and invasion. These results demonstrated that CDKN2A-AS1activated the BMP-SMAD signaling pathway by directly bind with SOSTDC1 to promote EOC tumor growth. CDKN2A-AS1/SOSTDC1 axis may provide a novel therapeutic strategy for EOC treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Proteins/metabolism , Carcinogenesis/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Progression , Ovarian Neoplasms/metabolism , RNA, Antisense , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Smad Proteins, Receptor-Regulated/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Down-Regulation/genetics , Female , Gene Knockdown Techniques , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Long Noncoding/genetics , Transfection , Up-Regulation/genetics
8.
J Cardiovasc Pharmacol ; 77(4): 480-490, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33818551

ABSTRACT

ABSTRACT: Atherosclerosis is a chronic lipid-induced inflammation of the vessel wall. Oxidized low-density lipoprotein was confirmed to drive the onset of atherogenesis. Zinc finger e-box-binding homeobox 1 antisense 1 (ZEB1-AS1) is a long noncoding RNA that is involved in human diseases, including atherosclerosis. In this study, the role of exosomes-mediated ZEB1-AS1 and its underlying mechanisms in atherosclerosis were explored in oxidized low-density lipoprotein (ox-LDL)-treated human umbilical vein endothelial cells (HUVECs). Exosomes were extracted from HUVECs. Quantitative real-time polymerase chain reaction was conducted to measure the expression of ZEB1-AS1, microRNA-590-5p (miR-590-5p), or erythroblastosis virus E26 oncogene homolog 1 (ETS1) in cells or exosomes. Cell proliferation and apoptosis were assessed by MTT assay and flow cytometry analysis, respectively. Western blot was performed to detect apoptosis-related factors, ETS1, and TGF-ß/Smad pathway protein levels. The secretion of inflammatory factors in supernatant was detected by ELISA assay. Oxidative stress damage indicators were used to assess cellular damage. Relationship between miR-590-5p and ZEB1-AS1 or ETS1 was analyzed. Our data indicated that ox-LDL-induced exosomes-mediated ZEB1-AS1 in HUVECs. Ox-LDL treatment resulted in limited proliferation, proapoptosis, inflammation, and oxidative stress damage, whereas knockdown of ZEB1-AS1 could reverse these effects. Mechanically, ZEB1-AS1 sponged miR-590-5p to regulate ETS1 expression. MiR-590-5p knockdown inverted effects above of si-ZEB1-AS1 on HUVECs under ox-LDL exposure. Moreover, ETS1 reversed miR-590-5p-induced effects and activated the TGF-ß/Smad pathway in ox-LDL-treated HUVECs. Taken together, our findings demonstrated that exosomes-mediated ZEB1-AS1 enhanced cell injuries by miR-590-5p/ETS1 axis through the TGF-ß/Smad pathway in ox-LDL-induced HUVECs, suggesting that inhibiting ZEB1-AS1 might be an effective way for atherosclerosis treatment.


Subject(s)
Atherosclerosis/metabolism , Exosomes/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Long Noncoding/metabolism , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Proliferation , Cells, Cultured , Exosomes/genetics , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Lipoproteins, LDL/toxicity , MicroRNAs/genetics , Phosphorylation , Proto-Oncogene Protein c-ets-1/genetics , RNA, Long Noncoding/genetics , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
9.
J Cell Physiol ; 236(11): 7533-7543, 2021 11.
Article in English | MEDLINE | ID: mdl-33844290

ABSTRACT

The epithelial-mesenchymal interactions are essential for the initiation and regulation of the development of teeth. Following the initiation of tooth development, numerous growth factors are secreted by the dental epithelium and mesenchyme that play critical roles in cellular differentiation. During tooth morphogenesis, the dental epithelial stem cells differentiate into several cell types, including inner enamel epithelial cells, which then differentiate into enamel matrix-secreting ameloblasts. Recently, we reported that the novel basic-helix-loop-helix transcription factor, AmeloD, is actively engaged in the development of teeth as a regulator of dental epithelial cell motility. However, the gene regulation mechanism of AmeloD is still unknown. In this study, we aimed to uncover the mechanisms regulating AmeloD expression during tooth development. By screening growth factors that are important in the early stages of tooth formation, we found that TGF-ß1 induced AmeloD expression and ameloblast differentiation in the dental epithelial cell line, SF2. TGF-ß1 phosphorylated ERK1/2 and Smad2/3 to induce AmeloD expression, whereas treatment with the MEK inhibitor, U0126, inhibited AmeloD induction. Promoter analysis of AmeloD revealed that the proximal promoter of AmeloD showed high activity in dental epithelial cell lines, which was enhanced following TGF-ß1 stimulation. These results suggested that TGF-ß1 activates AmeloD transcription via ERK1/2 phosphorylation. Our findings provide new insights into the mechanisms that govern tooth development.


Subject(s)
Ameloblasts/metabolism , Tooth Germ/metabolism , Transcription Factors, General/metabolism , Transcription, Genetic , Ameloblasts/drug effects , Animals , Cell Differentiation , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Developmental , Mice, Knockout , Morphogenesis , Phosphorylation , Rats , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Tooth Germ/cytology , Tooth Germ/drug effects , Transcription Factors, General/genetics , Transcription, Genetic/drug effects , Transforming Growth Factor beta1/pharmacology
10.
Respir Res ; 21(1): 290, 2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33138822

ABSTRACT

BACKGROUND: The poor understanding of pathogenesis in idiopathic pulmonary fibrosis (IPF) impaired development of effective therapeutic strategies. The aim of the current study is to investigate the roles of long non-coding RNA H19 (lncRNA H19) in the pulmonary inflammation and fibrosis of IPF. METHODS: Bleomycin was used to induce pulmonary inflammation and fibrosis in mice. The mRNAs and proteins expression in lung tissues was determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. H19 knockout (H19-/-) mice were generated by CRISPR/Cas9. RESULTS: The expression of H19 mRNA was up-regulated in fibrotic lungs patients with IPF as well as in lungs tissues that obtained from bleomycin-treated mice. H19-/- mice suppressed bleomycin-mediated pulmonary inflammation and inhibited the Il6/Stat3 signaling. H19 deficiency ameliorated bleomycin-induced pulmonary fibrosis and repressed the activation of TGF-ß/Smad and S1pr2/Sphk2 in the lungs of bleomycin-treated mice. CONCLUSIONS: Our data suggests that H19 is a profibrotic lncRNA and a potential therapeutic target for IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/prevention & control , Lung/metabolism , Pneumonia/prevention & control , RNA, Long Noncoding/metabolism , Animals , Bleomycin , Case-Control Studies , Cell Proliferation , Databases, Genetic , Disease Models, Animal , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-6/metabolism , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , RNA, Long Noncoding/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
11.
Sci Rep ; 10(1): 16492, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020537

ABSTRACT

Airway remodelling with subepithelial fibrosis, which abolishes the physiological functions of the bronchial wall, is a major issue in bronchial asthma. Human bronchial fibroblasts (HBFs) derived from patients diagnosed with asthma display in vitro predestination towards TGF-ß1-induced fibroblast-to-myofibroblast transition (FMT), a key event in subepithelial fibrosis. As commonly used anti-asthmatic drugs do not reverse the structural changes of the airways, and the molecular mechanism of enhanced asthma-related TGF-ß1-induced FMT is poorly understood, we investigated the balance between the profibrotic TGF-ß/Smad2/3 and the antifibrotic TGF-ß/Smad1/5/9 signalling pathways and its role in the myofibroblast formation of HBF populations derived from asthmatic and non-asthmatic donors. Our findings showed for the first time that TGF-ß-induced activation of the profibrotic Smad2/3 signalling pathway was enhanced, but the activation of the antifibrotic Smad1/5/(8)9 pathway by TGF-ß1 was significantly diminished in fibroblasts from asthmatic donors compared to those from their healthy counterparts. The impairment of the antifibrotic TGF-ß/Smad1/5/(8)9 pathway in HBFs derived from asthmatic donors was correlated with enhanced FMT. Furthermore, we showed that Smad1 silencing in HBFs from non-asthmatic donors increased the FMT potential in these cells. Additionally, we demonstrated that activation of antifibrotic Smad signalling via BMP7 or isoliquiritigenin [a small-molecule activator of the TGF-ß/Smad1/5/(8)9 pathway] administration prevents FMT in HBFs from asthmatic donors through downregulation of profibrotic genes, e.g., α-SMA and fibronectin. Our data suggest that influencing the balance between the antifibrotic and profibrotic TGF-ß/Smad signalling pathways using BMP7-mimetic compounds presents an unprecedented opportunity to inhibit subepithelial fibrosis during airway remodelling in asthma.


Subject(s)
Asthma/metabolism , Fibroblasts/metabolism , Myofibroblasts/metabolism , Signal Transduction/physiology , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism , Adult , Airway Remodeling/physiology , Bronchi/metabolism , Case-Control Studies , Cells, Cultured , Female , Humans , Male , Middle Aged
12.
Biol Direct ; 15(1): 16, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33028367

ABSTRACT

BACKGROUND: Amelogenesis imperfecta (AI) is a type of hereditary diseases that manifest defects in the formation or mineralization of enamel. Recently, it is reported that inactivation of FAM20C, a well-known Golgi casein kinase, caused AI. However, the mechanism of it is still unknown. The aim of this study was to explore the molecular mechanism of AI, which caused by ablation of FAM20C. RESULTS: In the Sox2-Cre;Fam20Cfl/fl (cKO) mouse, we found abnormal differentiation of ameloblasts, improper formation and mineralization of enamel, and downregulation of both mRNA and protein level of enamel matrix proteins, including amelogenin (AMEL), ameloblastin (AMBN) and enamelin (ENAM). The levels of BMP2, BMP4 and BMP7, the ligands of BMP signaling pathway, and phosphorylation of Smad1/5/8, the key regulators of BMP signaling pathway, were all decreased in the enamel matrix and the ameloblast of the cKO mice, respectively. The expression of cyclin-dependent kinase inhibitor (P21), muscle segment homeobox genes 2 (Msx2), which are the target genes of the BMP signaling pathway, and laminin 3, the downstream factor of Msx2, were all significantly decreased in the ameloblasts of the cKO mice compared to the control mice. CONCLUSION: the results of our study suggest that ablation of FAM20C leads to AI through inhibiting the Smad dependent BMP signaling pathway in the process of amelogenesis.


Subject(s)
Amelogenesis Imperfecta/genetics , Bone Morphogenetic Proteins/genetics , Calcium-Binding Proteins/genetics , Dental Enamel Proteins/genetics , Extracellular Matrix Proteins/genetics , Signal Transduction , Smad Proteins, Receptor-Regulated/genetics , Animals , Bone Morphogenetic Proteins/metabolism , Calcium-Binding Proteins/metabolism , Dental Enamel Proteins/metabolism , Down-Regulation , Extracellular Matrix Proteins/metabolism , Mice , Mice, Knockout , Smad Proteins, Receptor-Regulated/metabolism
13.
Nutrients ; 12(10)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33096661

ABSTRACT

Although drug therapies are available for postmenopausal osteoporosis, these drugs are not free of side effects and long-term adherence to them are low. A safe and effective nutritional approach to counter postmenopausal osteoporosis is an important research goal. We fed ovariectomized (OVX) Sprague-Dawley rats a diet supplemented with 1% or 2% green tomato extract (GTE). After 12 weeks, micro-computed tomography scans revealed that GTE supplementation effectively prevented distal femur bone loss. This prevention was due to improved bone formation and suppressed bone resorption as observed by the regulation of osteoblast and osteoclast activities. GTE supplementation also improved bone formation through Bmp2-Smad 1/5/8-Runx2 signaling, while bone resorption was regulated by the receptor activator of nuclear factor kappa-B (RANKL)/osteoprogeterin (OPG) pathway. These results suggest that GTE supplementation prevents severe postmenopausal bone loss by maintaining the regulation of bone homeostasis in OVX rats. GTE as a diet supplement might be a potential novel alternative for the prevention of postmenopausal osteoporosis.


Subject(s)
Osteoporosis, Postmenopausal/prevention & control , Plant Extracts/therapeutic use , Solanum lycopersicum , Animals , Bone Density/drug effects , Bone Morphogenetic Protein 2/metabolism , Bone Resorption/prevention & control , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Female , Humans , Solanum lycopersicum/chemistry , Osteogenesis/drug effects , Osteoprotegerin/metabolism , Ovariectomy , Phytotherapy , RANK Ligand/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Tomatine/analogs & derivatives , Tomatine/analysis , Weight Gain
14.
Elife ; 92020 07 07.
Article in English | MEDLINE | ID: mdl-32633716

ABSTRACT

Organ growth and size are finely tuned by intrinsic and extrinsic signaling molecules. In Drosophila, the BMP family member Dpp is produced in a limited set of imaginal disc cells and functions as a classic morphogen to regulate pattern and growth by diffusing throughout imaginal discs. However, the role of TGFß/Activin-like ligands in disc growth control remains ill-defined. Here, we demonstrate that Myoglianin (Myo), an Activin family member, and a close homolog of mammalian Myostatin (Mstn), is a muscle-derived extrinsic factor that uses canonical dSmad2-mediated signaling to regulate wing size. We propose that Myo is a myokine that helps mediate an allometric relationship between muscles and their associated appendages.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Imaginal Discs/growth & development , Smad Proteins, Receptor-Regulated/physiology , Transforming Growth Factor beta/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Muscles/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
15.
Biochem Biophys Res Commun ; 528(3): 545-553, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32505342

ABSTRACT

Diabetic cardiomyopathy (DCM) is one of the main causes of heart failure in patients with diabetes. Cardiac fibrosis caused by endothelial mesenchymal transformation (EndMT) plays an important role in the pathogenesis of DCM. NLRC5 is a recently discovered immune and inflammatory regulatory molecule in the NOD-like receptor family, and is involved in organ fibrosis. In this study, we found that the expression of NLRC5 was up-regulated in endothelial cells (ECs) and cardiac fibroblasts (CFs) in diabetes models both in vivo and in vitro. NLRC5 knockdown significantly inhibited high glucose-induced EndMT. In addition, NLRC5 deficiency inhibited the expression of phosphorylated Smad2/3 and the activation of EndMT-related transcription factors in ECs induced by high glucose. However, the effect of NLRC5 deficiency on CFs was not obvious. In summary, our results suggest that NLRC5 deficiency ameliorates cardiac fibrosis in DCM by inhibiting EndMT through Smad2/3 signaling pathway and related transcription factors. NLRC5 is likely to be a biomarker and therapeutic target of cardiac fibrosis in diabetic cardiomyopathy.


Subject(s)
Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Intracellular Signaling Peptides and Proteins/deficiency , Animals , Cell Transdifferentiation , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/prevention & control , Fibrosis , Gene Knockdown Techniques , Glucose/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism
16.
J Int Med Res ; 48(5): 300060520903612, 2020 May.
Article in English | MEDLINE | ID: mdl-32475187

ABSTRACT

OBJECTIVES: The objective was to observe the effects of Astragalus polysaccharides on diabetes and on regulation of the TGF-ß/Smad signaling pathway. METHODS: A type 2 diabetic rat model was established with a high-fat diet in combination with low-dose streptozotocin (35 mg/kg). Astragalus polysaccharides were applied as treatment intervention and changes in blood glucose and kidney morphology and function were assessed. RESULTS: Eight weeks after model establishment, kidney weight as a proportion of total weight (KW/TW) in the high-, medium-, and low-dose Astragalus polysaccharide groups was significantly lower than that in the model group, and the KW/TW value gradually decreased with increasing dose of polysaccharides in each treatment group. Fasting blood glucose in the low- and medium-dose Astragalus polysaccharide groups was numerically lower than that in the model group and fasting blood glucose in rats in the high-dose group was significantly lower than that in the model group. Levels of 24-hour urinary microalbumin, creatinine, blood urea nitrogen, collagens I, III, and IV, α-smooth muscle actin, transforming growth factor-ß1, and Smad3 in Astragalus polysaccharide groups (all doses) were significantly lower than those in the model group. CONCLUSIONS: Astragalus polysaccharide significantly improved blood glucose and protected kidney function in a rat diabetes model.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Kidney/drug effects , Plant Extracts/pharmacology , Animals , Astragalus Plant/metabolism , Blood Glucose/metabolism , Creatinine/blood , Diabetic Nephropathies/blood , Diet, High-Fat , Disease Models, Animal , Kidney/metabolism , Male , Polysaccharides/pharmacology , Rats , Rats, Sprague-Dawley , Retrospective Studies , Signal Transduction/drug effects , Smad Proteins, Receptor-Regulated/metabolism , Streptozocin/metabolism , Streptozocin/pharmacology , Transforming Growth Factor beta1/metabolism
17.
Development ; 147(10)2020 05 21.
Article in English | MEDLINE | ID: mdl-32345746

ABSTRACT

Synapses exhibit an astonishing degree of adaptive plasticity in healthy and disease states. We have investigated whether synapses also adjust to life stages imposed by novel developmental programs for which they were never molded by evolution. Under conditions in which Drosophila larvae are terminally arrested, we have characterized synaptic growth, structure and function at the neuromuscular junction (NMJ). Although wild-type larvae transition to pupae after 5 days, arrested third instar (ATI) larvae persist for 35 days, during which time NMJs exhibit extensive overgrowth in muscle size, presynaptic release sites and postsynaptic glutamate receptors. Remarkably, despite this exuberant growth, stable neurotransmission is maintained throughout the ATI lifespan through a potent homeostatic reduction in presynaptic neurotransmitter release. Arrest of the larval stage in stathmin mutants also reveals a degree of progressive instability and neurodegeneration that was not apparent during the typical larval period. Hence, an adaptive form of presynaptic depression stabilizes neurotransmission during an extended developmental period of unconstrained synaptic growth. More generally, the ATI manipulation provides a powerful system for studying neurodegeneration and plasticity across prolonged developmental timescales.


Subject(s)
Drosophila/growth & development , Drosophila/genetics , Larva/growth & development , Larva/genetics , Long-Term Synaptic Depression/genetics , Nerve Degeneration/genetics , Neuromuscular Junction/growth & development , Animals , Axons/pathology , Drosophila Proteins/genetics , Female , Homeostasis/genetics , Male , Mutation , Neuromuscular Junction/metabolism , RNA Interference , Smad Proteins, Receptor-Regulated/genetics , Stathmin/genetics , Synapses/metabolism , Synaptic Transmission/genetics
18.
Parasit Vectors ; 13(1): 164, 2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32245505

ABSTRACT

BACKGROUND: Smad proteins are essential cellular mediators within the transforming growth factor-ß (TGF-ß) superfamily. They directly transmit incoming signals from the cell surface receptors to the nucleus. In spite of their functional importance, almost nothing is known about Smad proteins in parasitic nematodes including Haemonchus contortus, an important blood-sucking nematode of small ruminants. METHODS: Based on genomic and transcriptome data for H. contortus and using bioinformatics methods, a Smad homologue (called Hco-daf-8) was inferred from H. contortus and the structural characteristics of this gene and its encoded protein Hco-DAF-8 established. Using real-time PCR and immunofluorescence assays, temporal transcriptional and spatial expression profiles of Hco-daf-8 were studied. Gene rescue in Caenorhabditis elegans was then applied to assess the function of Hco-daf-8 and a specific inhibitor of human Smad3 (called SIS3) was employed to evaluate the roles of Hco-DAF-8 in H. contortus development. RESULTS: The features of Hco-DAF-8 (502 amino acids), including conserved R-Smad domains and residues of the L3-loop that determine pathway specificity, are consistent with a TGF-ß type I receptor-activated R-Smad. The Hco-daf-8 gene was transcribed in all developmental stages of H. contortus studied, with a higher level of transcription in the fourth-stage larval (L4) females and the highest level in adult males. Hco-DAF-8 was expressed in the platymyarian muscular cells, intestine and reproductive system of adult stages. Gene rescue experiments showed that Hco-daf-8 was able to partially rescue gene function in a daf-8 deficient mutant strain of C. elegans, leading to a resumption of normal development. In H. contortus, SIS3 was shown to affect H. contortus development from the exsheathed third-stage larvae (L3s) to L4s in vitro. CONCLUSIONS: These findings suggest that Hco-DAF-8, encoded by the gene Hco-daf-8, is an important cellular mediator of H. contortus development via the TGF-ß signalling pathway. They provide a basis for future explorations of Hco-DAF-8 and associated pathways in H. contortus and other important parasitic nematodes.


Subject(s)
Haemonchus/genetics , Helminth Proteins/genetics , Smad Proteins, Receptor-Regulated/genetics , Transcriptome , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Computational Biology , Female , Gene Expression Profiling , Genomics , Haemonchus/growth & development , Male , Sequence Alignment , Signal Transduction , Smad Proteins, Receptor-Regulated/classification
19.
Biol Pharm Bull ; 43(3): 533-539, 2020.
Article in English | MEDLINE | ID: mdl-32115512

ABSTRACT

Renal interstitial fibrosis (RIF) is a common pathological characteristic associated with end-stage renal disease. However, treatment strategies for RIF are still very limited. In this study, we reported that kaempferol, a classic flavonoid, exhibited strong and widely inhibitory effect on the expression of fibrosis related genes in transforming growth factor beta 1 (TGF-ß1) treated NRK-52E cells. Further studies revealed that kaempferol inhibited TGF-ß1 induced epithelial-mesenchymal transition (EMT) process of NRK-52E cells and improved renal function deterioration and RIF in unilateral ureteral obstruction (UUO) rats. After exploring the underlying mechanisms, we found that kaempferol was able to activate the BMP-7-Smad1/5 pathway, rather than the TGF-ß1-Smad2/3 pathway. To further validate these results, DMH1 and BMP-7 knockdown were utilized at the cellular level and the results showed that both methods were able to antagonize the effects of kaempferol on the EMT process of NRK-52E cells induced by TGF-ß1. In UUO rats, inhibition of BMP-7 signaling by DMH1 also reversed the effects of kaempferol on renal function decline and RIF. Taken together, our findings demonstrated that kaempferol could be a good candidate for renal fibrosis treatment.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Kaempferols/pharmacology , Kidney Diseases/metabolism , Smad Proteins, Receptor-Regulated/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Animals , Cell Line , Collagen/metabolism , Epithelial Cells , Fibrosis , Kidney/drug effects , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
20.
Cardiovasc Drugs Ther ; 34(1): 41-52, 2020 02.
Article in English | MEDLINE | ID: mdl-32096002

ABSTRACT

PURPOSE: The pleiotropic roles of phosphodiesterase-5 inhibitors (PDE5is) in cardiovascular diseases have attracted attention. The effect of vardenafil (a PDE5i) is partly mediated through reduced oxidative stress, but it is unclear whether vardenafil protects against hydrogen peroxide (H2O2)-induced endothelial cell injury, and the molecular mechanisms that are involved remain unknown. We determined the protective role of vardenafil on H2O2-induced endothelial cell injury in cultured human umbilical vein endothelial cells (HUVECs). METHODS AND RESULTS: Vardenafil decreased the number of TUNEL-positive cells, increased the Bcl2/Bax ratio, and ameliorated the numbers of BrdU-positive cells in H2O2-treated HUVECs. The bone morphogenetic protein receptor (BMPR)/p-Smad/MSX2 pathway was enhanced in response to H2O2, and vardenafil treatment could normalize this pathway. To determine whether the BMP pathway is involved, we blocked the BMP pathway using dorsomorphin, which abolished the protective effects of vardenafil. We found that vardenafil improved the H2O2-induced downregulation of BMP-binding endothelial regulator protein (BMPER), which possibly intersects with the BMP pathway in the regulation of endothelial cell injury in response to oxidative stress. CONCLUSIONS: We demonstrated for the first time that exogenous H2O2 activates BMPR expression and promotes Smad1/5/8 phosphorylation. Additionally, vardenafil can attenuate H2O2-induced endothelial cell injury in HUVECs. Vardenafil decreases apoptosis through an improved Bcl-2/Bax ratio and increases cell proliferation. Vardenafil protects against endothelial cell injury through ameliorating the intracellular oxidative stress level and BMPER expression. The protective role of vardenafil on H2O2-induced endothelial cell injury is mediated through BMPR/p-Smad/MSX2 in HUVECs.


Subject(s)
Antioxidants/pharmacology , Bone Morphogenetic Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Smad Proteins, Receptor-Regulated/metabolism , Vardenafil Dihydrochloride/pharmacology , Apoptosis/drug effects , Bone Morphogenetic Protein Receptors/metabolism , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...