Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.101
Filter
1.
Bull Exp Biol Med ; 177(1): 35-38, 2024 May.
Article in English | MEDLINE | ID: mdl-38954301

ABSTRACT

The features of the participation of Smad3 in the functioning of neural stem cells (NSC), neuronal committed precursors (NCP), and neuroglial elements were studied in vitro. It was found that this intracellular signaling molecule enhances the clonogenic and proliferative activities of NCP and inhibits specialization of neuronal precursors. At the same time, Smad3 does not participate in the realization of the growth potential of NSC. With regard to the secretory function (production of neurotrophic growth factors) of neuroglial cells, the stimulating role of Smad3-mediated signaling was shown. These results indicate the promise of studying the possibility of using Smad3 as a fundamentally new target for neuroregenerative agents.


Subject(s)
Cell Proliferation , Neural Stem Cells , Neuroglia , Smad3 Protein , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Animals , Neuroglia/metabolism , Neuroglia/cytology , Cell Proliferation/physiology , Signal Transduction , Cell Differentiation/physiology , Cells, Cultured , Rats , Neurons/metabolism , Neurons/cytology , Mice
2.
Nat Commun ; 15(1): 5731, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977708

ABSTRACT

Neuropilin-1 (NRP1), a co-receptor for various cytokines, including TGF-ß, has been identified as a potential therapeutic target for fibrosis. However, its role and mechanism in renal fibrosis remains elusive. Here, we show that NRP1 is upregulated in distal tubular (DT) cells of patients with transplant renal insufficiency and mice with renal ischemia-reperfusion (I-R) injury. Knockout of Nrp1 reduces multiple endpoints of renal injury and fibrosis. We find that Nrp1 facilitates the binding of TNF-α to its receptor in DT cells after renal injury. This signaling results in a downregulation of lysine crotonylation of the metabolic enzyme Cox4i1, decreases cellular energetics and exacerbation of renal injury. Furthermore, by single-cell RNA-sequencing we find that Nrp1-positive DT cells secrete collagen and communicate with myofibroblasts, exacerbating acute kidney injury (AKI)-induced renal fibrosis by activating Smad3. Dual genetic deletion of Nrp1 and Tgfbr1 in DT cells better improves renal injury and fibrosis than either single knockout. Together, these results reveal that targeting of NRP1 represents a promising strategy for the treatment of AKI and subsequent chronic kidney disease.


Subject(s)
Acute Kidney Injury , Fibrosis , Mice, Knockout , Neuropilin-1 , Receptor, Transforming Growth Factor-beta Type I , Reperfusion Injury , Smad3 Protein , Neuropilin-1/metabolism , Neuropilin-1/genetics , Animals , Humans , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Male , Tumor Necrosis Factor-alpha/metabolism , Signal Transduction , Mice, Inbred C57BL , Kidney Tubules/pathology , Kidney Tubules/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Collagen/metabolism
3.
Funct Integr Genomics ; 24(4): 123, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992207

ABSTRACT

Hepatocellular carcinoma (HCC) is a common malignancy with a poor prognosis. It has been proven that long non-coding RNAs (lncRNAs) play an essential role in regulating HCC progression. However, the involvement of LINC01094 in regulating epithelial-mesenchymal transition (EMT) in HCC remains unclear. LINC01094 expression in HCC patients was retrieved from the Cancer Genome Atlas database. Overexpressing and downregulating LINC01094 were conducted to investigate its biological functions using Hep3B, SNU-387, and HuH-7 cells. Western blotting and morphological observation were performed to study the EMT in HCC cells. Transwell assay was adopted to determine the migration and invasion of HCC cells. The underlying mechanism of competitive endogenous RNAs (ceRNAs) was investigated using bioinformatics analysis, quantitative reverse-transcription polymerase chain reaction, and rescue experiments. Elevated LINC01094 expression was observed in HCC and associated with a poor prognosis. Knockdown of LINC01094 expression in SNU-387 and HuH-7 cells could inhibit migration, invasion, and EMT markers. Overexpression of LINC01094 indicated that LINC01094 promoted EMT via the TGF-ß/SMAD signaling pathway. The bioinformatics analysis revealed that miR-122-5p was a target of LINC01094. The miRWalk database analysis showed that TGFBR2, SMAD2, and SMAD3 were downstream targets of miR-122-5p. Mechanically, LINC01094 acted as a ceRNA that facilitated HCC metastasis by sponging miR-122-5p to regulate the expression of TGFBR2, SMAD2, and SMAD3. Further, TGF-ß1 could enhance the expression of LINC01094, forming a positive feedback loop. TGF-ß1-induced LINC01094 expression promotes HCC cell migration and invasion by targeting the miR-122-5p/TGFBR2-SMAD2-SMAD3 axis. LINC01094 may be a potential prognostic biomarker and therapeutic target for HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular , Epithelial-Mesenchymal Transition , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Receptor, Transforming Growth Factor-beta Type II , Smad3 Protein , Transforming Growth Factor beta1 , Humans , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Signal Transduction
4.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38960622

ABSTRACT

A pleiotropic immunoregulatory cytokine, TGF-ß, signals via the receptor-regulated SMADs: SMAD2 and SMAD3, which are constitutively expressed in normal cells. Here, we show that selective repression of SMAD3 induces cDC differentiation from the CD115+ common DC progenitor (CDP). SMAD3 was expressed in haematopoietic cells including the macrophage DC progenitor. However, SMAD3 was specifically down-regulated in CD115+ CDPs, SiglecH- pre-DCs, and cDCs, whereas SMAD2 remained constitutive. SMAD3-deficient mice showed a significant increase in cDCs, SiglecH- pre-DCs, and CD115+ CDPs compared with the littermate control. SMAD3 repressed the mRNA expression of FLT3 and the cDC-related genes: IRF4 and ID2. We found that one of the SMAD transcriptional corepressors, c-SKI, cooperated with phosphorylated STAT3 at Y705 and S727 to repress the transcription of SMAD3 to induce cDC differentiation. These data indicate that STAT3 and c-Ski induce cDC differentiation by repressing SMAD3: the repressor of the cDC-related genes during the developmental stage between the macrophage DC progenitor and CD115+ CDP.


Subject(s)
Cell Differentiation , Dendritic Cells , Interferon Regulatory Factors , STAT3 Transcription Factor , Smad3 Protein , Animals , Cell Differentiation/genetics , Dendritic Cells/metabolism , Dendritic Cells/cytology , Smad3 Protein/metabolism , Smad3 Protein/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Mice , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Mice, Knockout , Mice, Inbred C57BL , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Phosphorylation , Signal Transduction
5.
Allergol Immunopathol (Madr) ; 52(4): 91-96, 2024.
Article in English | MEDLINE | ID: mdl-38970271

ABSTRACT

Asthma is a widely prevalent chronic disease that brings great suffering to patients and may result in death if it turns severe. Jolkinolide B (JB) is one diterpenoid component separated from the dried roots of Euphorbia fischeriana Steud (Euphorbiaceae), and has anti--inflammatory, antioxidative, and antitumor properties. However, the detailed regulatory role and associated regulatory mechanism in the progression of asthma remain elusive. In this work, it was demonstrated that the extensive infiltration of bronchial inflammatory cells and the thickening of airway wall were observed in ovalbumin (OVA)-induced mice, but these impacts were reversed by JB (10 mg/kg) treatment, indicating that JB relieved the provocative symptoms in OVA-induced asthma mice. In addition, JB can control OVA-triggered lung function and pulmonary resistance. Moreover, JB attenuated OVA-evoked inflammation by lowering the levels of interleukin (IL)-4, IL-5, and IL-13. Besides, the activated nuclear factor kappa B (NF-κB) and transforming growth factor-beta-mothers against decapentaplegic homolog 3 (TGFß/smad3) pathways in OVA-induced mice are rescued by JB treatment. In conclusion, it was disclosed that JB reduced allergic airway inflammation and airway remodeling in asthmatic mice by modulating the NF-κB and TGFß/smad3 pathways. This work could offer new opinions on JB for lessening progression of asthma.


Subject(s)
Airway Remodeling , Asthma , Disease Models, Animal , Diterpenes , Mice, Inbred BALB C , NF-kappa B , Ovalbumin , Animals , Asthma/drug therapy , Asthma/immunology , Airway Remodeling/drug effects , Mice , Diterpenes/pharmacology , Diterpenes/administration & dosage , Diterpenes/therapeutic use , Ovalbumin/immunology , NF-kappa B/metabolism , Female , Transforming Growth Factor beta/metabolism , Cytokines/metabolism , Smad3 Protein/metabolism , Signal Transduction/drug effects , Humans , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Euphorbia/chemistry
6.
Chem Pharm Bull (Tokyo) ; 72(7): 669-675, 2024.
Article in English | MEDLINE | ID: mdl-39010213

ABSTRACT

Tendon injury is a prevalent orthopedic disease that currently lacks effective treatment. Galangin (GLN) is a vital flavonoid found abundantly in galangal and is known for its natural activity. This study aimed to investigate the GLN-mediated molecular mechanism of tendon-derived stem cells (TDSCs) in tendon repair. The TDSCs were characterized using alkaline phosphatase staining, alizarin red S staining, oil red O staining, and flow cytometry. The effect of GLN treatment on collagen deposition was evaluated using Sirius red staining and quantitative (q)PCR, while a Western bot was used to assess protein levels and analyze pathways. Results showed that GLN treatment not only increased the collagen deposition but also elevated the mRNA expression and protein levels of multiple tendon markers like collagen type I alpha 1 (COL1A1), decorin (DCN) and tenomodulin (TNMD) in TDSCs. Moreover, GLN was also found to upregulate the protein levels of transforming growth factor ß1 (TGF-ß1) and p-Smad3 to activate the TGF-ß1/Smad3 signaling pathway, while GLN mediated collagen deposition in TDSCs was reversed by LY3200882, a TGF-ß receptor inhibitor. The study concluded that GLN-mediated TDSCs enhanced tendon repair by activating the TGF-ß1/Smad3 signaling pathway, suggesting a novel therapeutic option in treating tendon repair.


Subject(s)
Flavonoids , Signal Transduction , Smad3 Protein , Stem Cells , Tendons , Transforming Growth Factor beta1 , Flavonoids/pharmacology , Flavonoids/chemistry , Transforming Growth Factor beta1/metabolism , Signal Transduction/drug effects , Animals , Smad3 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Tendons/cytology , Tendons/metabolism , Tendons/drug effects , Rats , Cells, Cultured , Rats, Sprague-Dawley , Tendon Injuries/drug therapy , Tendon Injuries/metabolism
7.
Nat Commun ; 15(1): 5680, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971819

ABSTRACT

Obesity shapes anti-tumor immunity through lipid metabolism; however, the mechanisms underlying how colorectal cancer (CRC) cells utilize lipids to suppress anti-tumor immunity remain unclear. Here, we show that tumor cell-intrinsic ATP6V0A1 drives exogenous cholesterol-induced immunosuppression in CRC. ATP6V0A1 facilitates cholesterol absorption in CRC cells through RAB guanine nucleotide exchange factor 1 (RABGEF1)-dependent endosome maturation, leading to cholesterol accumulation within the endoplasmic reticulum and elevated production of 24-hydroxycholesterol (24-OHC). ATP6V0A1-induced 24-OHC upregulates TGF-ß1 by activating the liver X receptor (LXR) signaling. Subsequently, the release of TGF-ß1 into the tumor microenvironment by CRC cells activates the SMAD3 pathway in memory CD8+ T cells, ultimately suppressing their anti-tumor activities. Moreover, we identify daclatasvir, a clinically used anti-hepatitis C virus (HCV) drug, as an ATP6V0A1 inhibitor that can effectively enhance the memory CD8+ T cell activity and suppress tumor growth in CRC. These findings shed light on the potential for ATP6V0A1-targeted immunotherapy in CRC.


Subject(s)
CD8-Positive T-Lymphocytes , Cholesterol , Colorectal Neoplasms , Signal Transduction , Transforming Growth Factor beta1 , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Animals , Cholesterol/metabolism , Mice , Cell Line, Tumor , Transforming Growth Factor beta1/metabolism , Immunologic Memory , Vacuolar Proton-Translocating ATPases/metabolism , Tumor Microenvironment/immunology , Liver X Receptors/metabolism , Hydroxycholesterols/metabolism , Hydroxycholesterols/pharmacology , Pyrrolidines/pharmacology , Smad3 Protein/metabolism , Mice, Inbred C57BL , Carbamates/pharmacology
8.
Curr Med Sci ; 44(3): 519-528, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38842774

ABSTRACT

OBJECTIVE: Intestinal fibrosis is a refractory complication of inflammatory bowel disease (IBD). Tumor necrosis factor ligand-related molecule-1A (TL1A) is important for IBD-related intestinal fibrosis in a dextran sodium sulfate (DSS)-induced experimental colitis model. This study aimed to explore the effects of TL1A on human colonic fibroblasts. METHODS: A trinitrobenzene sulfonic acid (TNBS)-induced experimental colitis model of LCK-CD2-TL1A-GFP transgenic (Tg) or wild-type (WT) mice was established to determine the effect and mechanism of TL1A on intestinal fibrosis. The human colonic fibroblast CCD-18Co cell line was treated concurrently with TL1A and human peripheral blood mononuclear cell (PBMC) supernatant. The proliferation and activation of CCD-18Co cells were detected by BrdU assays, flow cytometry, immunocytochemistry and Western blotting. Collagen metabolism was tested by Western blotting and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS: The level of collagen metabolism in the TNBS+ethyl alcohol (EtOH)/Tg group was greater than that in the TNBS+EtOH/WT group. Transforming growth factor-ß1 (TGF-ß1) and p-Smad3 in the TNBS+EtOH/Tg group were upregulated as compared with those in the TNBS+EtOH/WT group. The proliferation of CCD-18Co cells was promoted by the addition of human PBMC supernatant supplemented with 20 ng/mL TL1A, and the addition of human PBMC supernatant and TL1A increased CCD-18Co proliferation by 24.4% at 24 h. TL1A promoted cell activation and increased the levels of COL1A2, COL3A1, and TIMP-1 in CCD-18Co cells. Treatment of CCD-18Co cells with TL1A increased the expression of TGF-ß1 and p-Smad3. CONCLUSION: TL1A promotes TGF-ß1-mediated intestinal fibroblast activation, proliferation, and collagen deposition and is likely related to an increase in the TGF-ß1/Smad3 signaling pathway.


Subject(s)
Cell Proliferation , Fibroblasts , Fibrosis , Signal Transduction , Smad3 Protein , Transforming Growth Factor beta1 , Tumor Necrosis Factor Ligand Superfamily Member 15 , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Humans , Fibroblasts/metabolism , Fibroblasts/pathology , Animals , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Mice , Colon/metabolism , Colon/pathology , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/genetics , Cell Line , Mice, Transgenic , Trinitrobenzenesulfonic Acid , Disease Models, Animal , Leukocytes, Mononuclear/metabolism
9.
Int Immunopharmacol ; 136: 112375, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38823182

ABSTRACT

Lung fibrosis is a critical interstitial lung disease with poor prognosis. There is an urgent need to develop a proper and cost-effective therapeutic modality that can reverse and/or ameliorate lung fibrosis. Vitamin E is one of the widely investigated dietary antioxidants which has been linked to improvement of many health problems. The current study was conducted to evaluate the possible roles of vitamin E in prevention and treatment of bleomycin (BLM) induced lung fibrosis. Physiological, anatomical, histopathological and immunohistochemical studies were done to assess and compare between the structure and function of the lung tissue in lung fibrosis model, early and late treated groups with vitamin E. Furthermore, measurement of transforming growth factor-ß(TGF-ß), E-cadherin, Smad-3, BAX, BCL2, malondialdehyde (MDA), and superoxide dismutase (SOD) were done. The study revealed that administration of vitamin E helped to improve signs of lung fibrosis, as reflected by amelioration of structure and functions of lungs as well as the decrease in TGF-ß levels and inhibition of α-SMA/collagen I profibrotic pathway. These findings highlight the importance of administration of vitamin E as a prophylactic agent prior to BLM therapy and as an adjuvant treatment in cases of lung fibrosis.


Subject(s)
Antioxidants , Bleomycin , Lung , Pulmonary Fibrosis , Transforming Growth Factor beta , Vitamin E , Animals , Vitamin E/therapeutic use , Vitamin E/pharmacology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Lung/pathology , Lung/drug effects , Rats , Transforming Growth Factor beta/metabolism , Male , Antioxidants/therapeutic use , Antioxidants/pharmacology , Smad3 Protein/metabolism , Superoxide Dismutase/metabolism , Malondialdehyde/metabolism , Cadherins/metabolism , Rats, Wistar , Actins/metabolism , Disease Models, Animal , Humans
10.
Nat Commun ; 15(1): 4758, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902234

ABSTRACT

To uncover molecular changes underlying blood-brain-barrier dysfunction in Alzheimer's disease, we performed single nucleus RNA sequencing in 24 Alzheimer's disease and control brains and focused on vascular and astrocyte clusters as main cell types of blood-brain-barrier gliovascular-unit. The majority of the vascular transcriptional changes were in pericytes. Of the vascular molecular targets predicted to interact with astrocytic ligands, SMAD3, upregulated in Alzheimer's disease pericytes, has the highest number of ligands including VEGFA, downregulated in Alzheimer's disease astrocytes. We validated these findings with external datasets comprising 4,730 pericyte and 150,664 astrocyte nuclei. Blood SMAD3 levels are associated with Alzheimer's disease-related neuroimaging outcomes. We determined inverse relationships between pericytic SMAD3 and astrocytic VEGFA in human iPSC and zebrafish models. Here, we detect vast transcriptome changes in Alzheimer's disease at the gliovascular-unit, prioritize perturbed pericytic SMAD3-astrocytic VEGFA interactions, and validate these in cross-species models to provide a molecular mechanism of blood-brain-barrier disintegrity in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Astrocytes , Blood-Brain Barrier , Pericytes , Smad3 Protein , Vascular Endothelial Growth Factor A , Zebrafish , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Humans , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Astrocytes/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Pericytes/metabolism , Pericytes/pathology , Male , Induced Pluripotent Stem Cells/metabolism , Female , Aged , Transcriptome , Brain/metabolism , Brain/pathology , Brain/blood supply , Aged, 80 and over , Disease Models, Animal
11.
Open Biol ; 14(6): 230427, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862020

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a monogenic cardiac disorder commonly induced by sarcomere gene mutations. However, the mechanism for HCM is not well defined. Here, we generated transgenic MYH7 R453C and MYH6 R453C piglets and found both developed typical cardiac hypertrophy. Unexpectedly, we found serious fibrosis and cardiomyocyte loss in the ventricular of MYH7 R453C, not MYH6 R453C piglets, similar to HCM patients. Then, RNA-seq analysis and western blotting identified the activation of ERK1/2 and PI3K-Akt pathways in MYH7 R453C. Moreover, we observed an increased expression of fetal genes and an excess of reactive oxygen species (ROS) in MYH7 R453C piglet models, which was produced by Nox4 and subsequently induced inflammatory response. Additionally, the phosphorylation levels of Smad2/3, ERK1/2 and NF-kB p65 proteins were elevated in cardiomyocytes with the MYH7 R453C mutation. Furthermore, epigallocatechin gallate, a natural bioactive compound, could be used as a drug to reduce cell death by adjusting significant downregulation of the protein expression of Bax and upregulated Bcl-2 levels in the H9C2 models with MYH7 R453C mutation. In conclusion, our study illustrated that TGF-ß/Smad2/3, ERK1/2 and Nox4/ROS pathways have synergistic effects on cardiac remodelling and inflammation in MYH7 R453C mutation.


Subject(s)
Myosin Heavy Chains , NADPH Oxidase 4 , NF-kappa B , Reactive Oxygen Species , Signal Transduction , Transforming Growth Factor beta , Animals , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/genetics , Transforming Growth Factor beta/metabolism , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Swine , Myocytes, Cardiac/metabolism , Humans , Cardiac Myosins/metabolism , Cardiac Myosins/genetics , Disease Models, Animal , MAP Kinase Signaling System , Animals, Genetically Modified , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mutation , Smad3 Protein/metabolism , Smad3 Protein/genetics , Ventricular Remodeling , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Rats
12.
Sci Rep ; 14(1): 12716, 2024 06 03.
Article in English | MEDLINE | ID: mdl-38830933

ABSTRACT

To explore the molecular pathogenesis of pulmonary arterial hypertension (PAH) and identify potential therapeutic targets, we performed transcriptome sequencing of lung tissue from mice with hypoxia-induced pulmonary hypertension. Our Gene Ontology analysis revealed that "extracellular matrix organization" ranked high in the biological process category, and matrix metallopeptidases (MMPs) and other proteases also played important roles in it. Moreover, compared with those in the normoxia group, we confirmed that MMPs expression was upregulated in the hypoxia group, while the hub gene Timp1 was downregulated. Crocin, a natural MMP inhibitor, was found to reduce inflammation, decrease MMPs levels, increase Timp1 expression levels, and attenuate hypoxia-induced pulmonary hypertension in mice. In addition, analysis of the cell distribution of MMPs and Timp1 in the human lung cell atlas using single-cell RNAseq datasets revealed that MMPs and Timp1 are mainly expressed in a population of fibroblasts. Moreover, in vitro experiments revealed that crocin significantly inhibited myofibroblast proliferation, migration, and extracellular matrix deposition. Furthermore, we demonstrated that crocin inhibited TGF-ß1-induced fibroblast activation and regulated the pulmonary arterial fibroblast MMP2/TIMP1 balance by inhibiting the TGF-ß1/Smad3 signaling pathway. In summary, our results indicate that crocin attenuates hypoxia-induced pulmonary hypertension in mice by inhibiting TGF-ß1-induced myofibroblast activation.


Subject(s)
Carotenoids , Hypertension, Pulmonary , Hypoxia , Matrix Metalloproteinase 2 , Tissue Inhibitor of Metalloproteinase-1 , Animals , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Mice , Hypoxia/metabolism , Hypoxia/complications , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Carotenoids/pharmacology , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Male , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Disease Models, Animal , Cell Proliferation/drug effects , Mice, Inbred C57BL , Smad3 Protein/metabolism , Cell Movement/drug effects , Lung/pathology , Lung/metabolism , Lung/drug effects
13.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 384-389, 2024 Jun 18.
Article in Chinese | MEDLINE | ID: mdl-38864121

ABSTRACT

OBJECTIVE: To explore the association between polymorphisms of transforming growth factor-ß (TGF-ß) signaling pathway and non-syndromic cleft lip with or without cleft palate (NSCL/P) among Asian populations, while considering gene-gene interaction and gene-environment interaction. METHODS: A total of 1 038 Asian NSCL/P case-parent trios were ascertained from an international consortium, which conducted a genome-wide association study using a case-parent trio design to investigate the genes affec-ting risk to NSCL/P. After stringent quality control measures, 343 single nucleotide polymorphism (SNP) spanning across 10 pivotal genes in the TGF-ß signaling pathway were selected from the original genome-wide association study(GWAS) dataset for further analysis. The transmission disequilibrium test (TDT) was used to test for SNP effects. The conditional Logistic regression models were used to test for gene-gene interaction and gene-environment interaction. Environmental factors collected for the study included smoking during pregnancy, passive smoking during pregnancy, alcohol intake during pregnancy, and vitamin use during pregnancy. Due to the low rates of exposure to smoking during pregnancy and alcohol consumption during pregnancy (<3%), only the interaction between maternal smoking during pregnancy and multivitamin supplementation during pregnancy was analyzed. The threshold for statistical significance was rigorously set at P =1.46×10-4, applying Bonferroni correction to account for multiple testing. RESULTS: A total of 23 SNPs in 4 genes yielded nominal association with NSCL/P (P<0.05), but none of these associations was statistically significant after Bonferroni' s multiple test correction. However, there were 6 pairs of SNPs rs4939874 (SMAD2) and rs1864615 (TGFBR2), rs2796813 (TGFB2) and rs2132298 (TGFBR2), rs4147358 (SMAD3) and rs1346907 (TGFBR2), rs4939874 (SMAD2) and rs1019855 (TGFBR2), rs4939874 (SMAD2) and rs12490466 (TGFBR2), rs2009112 (TGFB2) and rs4075748 (TGFBR2) showed statistically significant SNP-SNP interaction (P<1.46×10-4). In contrast, the analysis of gene-environment interactions did not yield any significant results after being corrected by multiple testing. CONCLUSION: The comprehensive evaluation of SNP associations and interactions within the TGF-ß signaling pathway did not yield any direct associations with NSCL/P risk in Asian populations. However, the significant gene-gene interactions identified suggest that the genetic architecture influencing NSCL/P risk may involve interactions between genes within the TGF-ß signaling pathway. These findings underscore the necessity for further investigations to unravel these results and further explore the underlying biological mechanisms.


Subject(s)
Cleft Lip , Cleft Palate , Gene-Environment Interaction , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Signal Transduction , Transforming Growth Factor beta , Humans , Cleft Palate/genetics , Cleft Lip/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Female , Asian People/genetics , Pregnancy , Male , Genetic Predisposition to Disease , Smad3 Protein/genetics , Risk Factors , Smad2 Protein/genetics , Smad2 Protein/metabolism , Epistasis, Genetic , Tobacco Smoke Pollution/adverse effects , Alcohol Drinking/genetics
14.
J Hazard Mater ; 474: 134823, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38852254

ABSTRACT

Nanoplastics (NPs) pollution has become a global environmental problem, raising numerous health concerns. However, the cardiotoxicity of NPs exposure and the underlying mechanisms have been understudied to date. To address this issue, we comprehensively evaluated the cardiotoxicity of polystyrene nanoplastics (PS-NPs) in both healthy and pathological states. Briefly, mice were orally exposed to four different concentrations (0 mg/day, 0.1 mg/day, 0.5 mg/day, and 2.5 mg/day) of 100-nm PS-NPs for 6 weeks to assess their cardiotoxicity in a healthy state. Considering that individuals with underlying health conditions are more vulnerable to the adverse effects of pollution, we further investigated the cardiotoxic effects of PS-NPs on pathological states induced by isoprenaline. Results showed that PS-NPs induced cardiomyocyte apoptosis, cardiac fibrosis, and myocardial dysfunction in healthy mice and exacerbated cardiac remodeling in pathological states. RNA sequencing revealed that PS-NPs significantly upregulated homeodomain interacting protein kinase 2 (HIPK2) in the heart and activated the P53 and TGF-beta signaling pathways. Pharmacological inhibition of HIPK2 reduced P53 phosphorylation and inhibited the activation of the TGF-ß1/Smad3 pathway, which in turn decreased PS-NPs-induced cardiotoxicity. This study elucidated the potential mechanisms underlying PS-NPs-induced cardiotoxicity and underscored the importance of evaluating nanoplastics safety, particularly for individuals with pre-existing heart conditions.


Subject(s)
Cardiotoxicity , Polystyrenes , Protein Serine-Threonine Kinases , Smad3 Protein , Transforming Growth Factor beta1 , Tumor Suppressor Protein p53 , Up-Regulation , Animals , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Cardiotoxicity/etiology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Polystyrenes/toxicity , Up-Regulation/drug effects , Male , Signal Transduction/drug effects , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Apoptosis/drug effects , Mice, Inbred C57BL , Nanoparticles/toxicity , Myocardium/metabolism , Myocardium/pathology
15.
PeerJ ; 12: e17485, 2024.
Article in English | MEDLINE | ID: mdl-38854800

ABSTRACT

Background: Cisplatin is a commonly used nephrotoxic drug and can cause acute kidney injury (AKI). In the present study, isobaric tags for relative and absolute quantification (iTRAQ) and parallel reaction monitoring (PRM)-based comparative proteomics were used to analyze differentially expressed proteins (DEPs) to determine the key molecular mechanism in mice with cisplatin-induced AKI in the presence or absence of SIS3, a specific p-smad3 inhibitor, intervention. Methods: The cisplatin-induced AKI mouse model was established and treated with SIS3. We used iTRAQ to search for DEPs, PRM to verify key DEPs and combined Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for bioinformatics analysis. We then assessed lipid deposition, malondialdehyde (MDA) and reactive oxygen species (ROS) and detected the expression of SREBF1, SCD1, CPT1A, PPARα and NDRG1 in vitro. Results: Proteomic analysis showed that the identified DEPs were mainly enriched in energy metabolism pathways, especially in lipid metabolism. When SIS3 was applied to inhibit the phosphorylation of Smad3, the expression of NDRG1 and fatty acid oxidation key proteins CPT1A and PPARα increased, the expression of lipid synthesis related proteins SREBF1 and SCD1 decreased and the production of lipid droplets, MDA and ROS decreased. Conclusion: SIS3 alleviates oxidative stress, reduces lipid accumulation and promotes fatty acid oxidation through NDRG1 in cisplatin-induced AKI. Our study provides a new candidate protein for elucidating the molecular mechanisms of fatty acid metabolism disorders in cisplatin-induced acute kidney injury.


Subject(s)
Acute Kidney Injury , Cisplatin , Proteomics , Cisplatin/adverse effects , Cisplatin/toxicity , Animals , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Proteomics/methods , Mice , Disease Models, Animal , Male , Smad3 Protein/metabolism , Smad3 Protein/genetics , Lipid Metabolism/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity
16.
Mol Med Rep ; 30(2)2024 08.
Article in English | MEDLINE | ID: mdl-38904199

ABSTRACT

Septic acute kidney injury (AKI) is considered as a severe and frequent complication that occurs during sepsis. Mounting evidence has confirmed the pivotal pathogenetic roles of microRNA (miRNA or miR) in sepsis­induced AKI; however, the role of miRNAs and their underlying mechanisms in sepsis­induced AKI have not been entirely understood. The present study aimed to elucidate the functions of special miRNAs during sepsis­induced AKI and its underlying mechanism. First, a number of differently expressed miRNAs was identified based on the microarray dataset GSE172044. Subsequently, lipopolysaccharide (LPS) was used to induce AKI in mice, and the role of miR­17­5p on AKI was clarified. Finally, the related molecular mechanisms were further examined by western blotting and immunohistochemical analysis. MiR­17­5p was found to be continuously decreased and reached the bottom at h 24 after AKI in mice. Functionally, injection of agomiR­17­5p could observably improve renal injury and survival rate, as well as inhibit inflammatory cytokine production and renal cell apoptosis in mice after AKI. On the contrary, injection of antagomiR­17­5p aggravated LPS­induced renal injury, inflammation and apoptosis in mice after AKI. Moreover, transforming growth factor ß receptor 2 (TGFßR2) was identified as a direct target of miR­17­5p, and its downstream phosphorylated Smad3 was also suppressed by miR­17­5p upregulation. Taken together, these results demonstrated that miR­17­5p overexpression may exhibit a beneficial effect by attenuating LPS­induced inflammation and apoptosis via regulating the TGFßR2/TGF­ß/Smad3 signaling pathway, indicating that miR­17­5p could act as a potential target for sepsis treatment.


Subject(s)
Acute Kidney Injury , Apoptosis , Inflammation , MicroRNAs , Receptor, Transforming Growth Factor-beta Type II , Sepsis , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Acute Kidney Injury/genetics , Sepsis/complications , Sepsis/metabolism , Sepsis/genetics , Apoptosis/genetics , Mice , Inflammation/genetics , Inflammation/metabolism , Male , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Lipopolysaccharides , Disease Models, Animal , Signal Transduction , Smad3 Protein/metabolism , Smad3 Protein/genetics , Mice, Inbred C57BL , Cytokines/metabolism
17.
Eur J Pharmacol ; 977: 176711, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38839029

ABSTRACT

Histone deacetylase (HDAC) inhibitors are potential candidates for treating pulmonary fibrosis. MPT0E028, a novel pan-HDAC inhibitor, has been reported to exhibit antitumor activity in several cancer cell lines. In this study, we investigated the mechanism underlying the inhibitory effects of MPT0E028 on the expression of fibrogenic proteins in human lung fibroblasts (WI-38). Our results revealed that MPT0E028 inhibited transforming growth factor-ß (TGF-ß)-, thrombin-, and endothelin 1-induced connective tissue growth factor (CTGF) expression in a concentration-dependent manner. In addition, MPT0E028 suppressed TGF-ß-stimulated expression of fibronectin, collagen I, and α-smooth muscle actin (α-SMA). Furthermore, MPT0E028 inhibited the TGF-ß-induced phosphorylation of c-Jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinase (ERK). MPT0E028 reduced the increase in SMAD3 and c-Jun phosphorylation, and SMAD3-and activator protein-1 (AP-1)-luciferase activities under TGF-ß stimulation. Transfection with mitogen-activated protein kinase phosphatase-1 (MKP-1) siRNA reversed the suppressive effects of MPT0E028 on TGF-ß-induced increases in CTGF expression; JNK, p38, and ERK phosphorylation; and SMAD3 and AP-1 activation. Moreover, MPT0E028 increased MKP-1 acetylation and activity in WI-38 cells. Pretreatment with MPT0E028 reduced the fibrosis score and fibronectin, collagen, and α-SMA expression in bleomycin-induced pulmonary fibrosis mice. In conclusion, MPT0E028 induced MKP-1 acetylation and activation, which in turn inhibited TGF-ß-stimulated JNK, p38, and ERK phosphorylation; SMAD3 and AP-1 activation; and subsequent CTGF expression in human lung fibroblasts. Thus, MPT0E028 may be a potential drug for treating pulmonary fibrosis.


Subject(s)
Connective Tissue Growth Factor , Dual Specificity Phosphatase 1 , Fibroblasts , Histone Deacetylase Inhibitors , Lung , Pulmonary Fibrosis , Transforming Growth Factor beta , Connective Tissue Growth Factor/metabolism , Humans , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/drug therapy , Animals , Histone Deacetylase Inhibitors/pharmacology , Mice , Lung/drug effects , Lung/pathology , Lung/cytology , Lung/metabolism , Transforming Growth Factor beta/metabolism , Dual Specificity Phosphatase 1/metabolism , Dual Specificity Phosphatase 1/genetics , Cell Line , Smad3 Protein/metabolism , Phosphorylation/drug effects , Male , Enzyme Activation/drug effects , Mice, Inbred C57BL
18.
Mol Biol Rep ; 51(1): 728, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861185

ABSTRACT

INTRODUCTION: Colorectal cancer (CRC) is the second common cancer and the fourth major reason of cancer death worldwide. Dysregulation of intracellular pathways, such as TGF-ß/SMAD signaling, contributes to CRC development. MicroRNAs (miRNAs) are post-transcriptional regulators that are involved in CRC pathogenesis. Here, we aimed to investigate the effect of miR-3613-3p on the TGF-ß /SMAD signaling pathway in CRC. METHODS & RESULTS: Bioinformatics analysis suggested that miR-3613-3p is a regulator of TGF-Β signaling downstream genes. Then, miR-3613-3p overexpression was followed by downregulation of TGF-ßR1, TGF-ßR2, and SMAD2 expression levels, detected by RT-qPCR. Additionally, dual luciferase assay supported the direct interaction of miR-3613-3p with 3'UTR sequences of TGF-ßR1 and TGF-ßR2 genes. Furthermore, reduced SMAD3 protein level following the miR-3613-3p overexpression verified its suppressive effect against TGF-ß signaling in HCT-116 cells, detected by western blot analysis. Finally, miR-3613-3p overexpression induced sub-G1 arrest in HCT116 cells, detected by flow cytometry, and promoted downregulation of cyclin D1 protein expression, which was detected by western blotting analysis. CONCLUSION: Our findings indicated that miR-3613-3p plays an important role in CRC by targeting the TGF-ß/SMAD signaling pathway and could be considered as a new candidate for further therapy investigations.


Subject(s)
Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , MicroRNAs , Signal Transduction , Transforming Growth Factor beta , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Smad2 Protein/metabolism , Smad2 Protein/genetics , Cell Proliferation/genetics , 3' Untranslated Regions/genetics , Cell Line, Tumor , Smad3 Protein/genetics , Smad3 Protein/metabolism
19.
FASEB J ; 38(11): e23729, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38847786

ABSTRACT

Diabetic nephropathy (DN) is one of the common microvascular complications in diabetic patients. Marrow mesenchymal stem cells (MSCs) have attracted attention in DN therapy but the underlying mechanism remains unclear. Here, we show that MSC administration alleviates high glucose (HG)-induced human kidney tubular epithelial cell (HK-2 cell) injury and ameliorates renal injury in DN mice. We identify that Smad2/3 is responsible for MSCs-regulated DN progression. The activity of Smad2/3 was predominantly upregulated in HG-induced HK-2 cell and DN mice and suppressed with MSC administration. Activation of Smad2/3 via transforming growth factor-ß1 (TGF-ß1) administration abrogates the protective effect of MSCs on HG-induced HK-2 cell injury and renal injury of DN mice. Smad2/3 has been reported to interact with methyltransferase of N6-methyladenosine (m6A) complex and we found a methyltransferase, Wilms' tumor 1-associating protein (WTAP), is involved in MSCs-Smad2/3-regulated DN development. Moreover, WTAP overexpression abrogates the improvement of MSCs on HG-induced HK-2 cell injury and renal injury of DN mice. Subsequently, α-enolase (ENO1) is the downstream target of WTAP-mediated m6A modification and contributes to the MSCs-mediated regulation. Collectively, these findings reveal a molecular mechanism in DN progression and indicate that Smad2/3/WTAP/ENO1 may present a target for MSCs-mediated DN therapy.


Subject(s)
Diabetic Nephropathies , Mesenchymal Stem Cells , Smad2 Protein , Smad3 Protein , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Animals , Mesenchymal Stem Cells/metabolism , Smad2 Protein/metabolism , Mice , Humans , Smad3 Protein/metabolism , Male , Mice, Inbred C57BL , Adenosine/metabolism , Adenosine/analogs & derivatives , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Signal Transduction , Methyltransferases/metabolism , Methyltransferases/genetics , Mesenchymal Stem Cell Transplantation/methods , Transforming Growth Factor beta1/metabolism , Cell Line
20.
Arch Dermatol Res ; 316(7): 338, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847916

ABSTRACT

Diabetic foot ulcer (DFU) is a predominant complication of diabetes mellitus with poor prognosis accompanied by high amputation and mortality rates. Dang-Gui-Si-Ni decoction (DSD), as a classic formula with a long history in China, has been found to improve DFU symptoms. However, mechanism of DSD for DFU therapy remains unclear with no systematic elaboration. In vivo, following establishment of DFU rat model, DSD intervention with low, medium and high doses was done, with Metformin (DM) as a positive control group. With wound healing detection, pathological changes by HE staining, inflammatory factor expression by ELISA and qRT-PCR, oxidative stress levels by ELISA, and AGEs/RAGE/TGF-ß/Smad2/3 expression by Western blot were performed. In vitro, intervention with LY2109761 (TGF-ß pathway inhibitor) based on DSD treatment in human dermal fibroblast-adult (HDF-a) cells was made. Cell viability by CCK8, migration ability by cell scratch, apoptosis by flow cytometry, and AGEs/RAGE/TGF-ß/Smad2/3 expression by Western blot were measured. DFU rats exhibited elevated AGEs/RAGE expression, whereas decreased TGF-ß1 and p-Smad3/Smad3 protein expression, accompanied by higher IL-1ß, IL-6, TNF-α levels, and oxidative stress. DSD intervention reversed above effects. Glucose induction caused lower cell viability, migration, TGF-ß1 and p-Smad3/Smad3 protein expression, with increased apoptosis and AGEs/RAGE expression in HDF-a cells. These effects were reversed after DSD intervention, and further LY2109761 intervention inhibited DSD effects in cells. DSD intervention may facilitate wound healing in DFU by regulating expression of AGEs/RAGE/TGF-ß/Smad2/3, providing scientific experimental evidence for DSD clinical application for DFU therapy.


Subject(s)
Diabetic Foot , Drugs, Chinese Herbal , Glycation End Products, Advanced , Smad2 Protein , Smad3 Protein , Wound Healing , Diabetic Foot/drug therapy , Diabetic Foot/metabolism , Diabetic Foot/pathology , Animals , Wound Healing/drug effects , Rats , Drugs, Chinese Herbal/pharmacology , Smad2 Protein/metabolism , Humans , Smad3 Protein/metabolism , Glycation End Products, Advanced/metabolism , Male , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Rats, Sprague-Dawley , Oxidative Stress/drug effects , Apoptosis/drug effects , Disease Models, Animal , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Fibroblasts/metabolism , Fibroblasts/drug effects , Cell Movement/drug effects , Cell Survival/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...