Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 661
Filter
1.
Bioorg Chem ; 147: 107374, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636433

ABSTRACT

The incidence of idiopathic pulmonary fibrosis (IPF) has been steadily increasing each year, posing significant challenges in its treatment. In this study, we conducted the design and synthesis of 23 new inhibitors that specifically target the TGF-ß1/Smad3 pathway. Initially, we employed a cell model of TGF-ß-induced pulmonary fibrosis, using cell survival rate and HYP expression as indicators to identify the potent ingredient 5aa, which demonstrated significant anti-pulmonary fibrosis activity. Subsequently, we induced mice with bleomycin (BLM) to establish an experimental animal model of pulmonary fibrosis, and evaluated the pharmacodynamics of 5aa in vivo against pulmonary fibrosis. The alterations in HYP and collagen levels in BLM-induced pulmonary fibrosis mice were analyzed using ELISA and immunohistochemistry techniques. The results indicated that compound 5aa effectively suppressed the fibrotic response induced by TGF-ß1, inhibited the expression of the fibrotic marker α-SMA, and hindered the EMT process in NIH3T3 cells. Additionally, oral administration of 5aa demonstrated significant therapeutic effects in a mouse model of IPF, comparable to the established drug Nintedanib. Moreover, compound 5aa exhibited higher bioavailability in vivo compared to Nintedanib. These collective outcomes suggest that 5aa holds promise as a potential inhibitor of TGF-ß1/Smad3 signaling for the treatment of IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Signal Transduction , Smad3 Protein , Transforming Growth Factor beta1 , Animals , Smad3 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/chemically induced , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Mice , Signal Transduction/drug effects , Molecular Structure , Humans , Bleomycin , Structure-Activity Relationship , Mice, Inbred C57BL , NIH 3T3 Cells , Dose-Response Relationship, Drug , Male
2.
Eur J Med Chem ; 271: 116395, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38626523

ABSTRACT

The transforming growth factor ß1 (TGFß1)/SMAD signaling pathway regulates many vital physiological processes. The development of potent inhibitors targeting activin receptor-like kinase 5 (ALK5) would provide potential treatment reagents for various diseases. A significant number of ALK5 inhibitors have been discovered, and they are currently undergoing clinical evaluation at various stages. However, the clinical demands were far from being met. In this study, we utilized an alternative conformation-similarity-based virtual screening (CSVS) combined with a fragment-based drug designing (FBDD) strategy to efficiently discover a potent and active hit with a novel chemical scaffold. After structural optimization in the principle of group replacement, compound 57 was identified as the most promising ALK5 inhibitor. Compound 57 demonstrated significant inhibitory effects against the TGF-ß1/SMAD signaling pathway. It could markedly attenuate the production of extracellular matrix (ECM) and deposition of collagen. Also, the lead compound showed adequate pharmacokinetic (PK) properties and good in vivo tolerance. Moreover, treatment with compound 57 in two different xerograph models showed significant inhibitory effects on the growth of pancreatic cancer cells. These results suggested that lead compound 57 refers as a promising ALK5 inhibitor both in vitro and in vivo, which merits further validation.


Subject(s)
Drug Design , Protein Kinase Inhibitors , Pyrazoles , Pyrimidines , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Humans , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Animals , Molecular Structure , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Mice , Cell Line, Tumor , Drug Screening Assays, Antitumor , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism
3.
Adv Mater ; 36(15): e2304328, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38229577

ABSTRACT

Currently, certain cancer patients exhibit resistance to radiotherapy due to reduced DNA damage under hypoxic conditions and acquired immune tolerance triggered by transforming growth factor-ß1 (TGF-ß1) and membrane-localized programmed death ligand-1 (PD-L1). Meanwhile, cytoplasm-distributed PD-L1 induces radiotherapy resistance through accelerating DNA damage repair (DDR). However, the disability of clinically used PD-L1 antibodies in inhibiting cytoplasm-distributed PD-L1 limits their effectiveness. Therefore, a nanoadjuvant is developed to sensitize cancer to radiotherapy via multi-level immunity activation through depressing PD-L1 and TGF-ß1 by triphenylphosphine-derived metformin, and activating the cGAS-STING pathway by generating Mn2+ from MnO2 and producing more dsDNA via reversing tumor hypoxia and impairing DDR. Thus, Tpp-Met@MnO2@Alb effectively enhances the efficiency of radiotherapy to inhibit the progression of irradiated local and abscopal tumors and tumor lung metastases, offering a long-term memory of antitumor immunity without discernible side effects. Overall, Tpp-Met@MnO2@Alb has the potential to be clinically applied for overcoming radio-immunotherapy resistance.


Subject(s)
Adjuvants, Pharmaceutic , Lung Neoplasms , Neoplasms , Humans , B7-H1 Antigen/antagonists & inhibitors , Immunotherapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Manganese Compounds/pharmacology , Neoplasms/radiotherapy , Neoplasms/therapy , Oxides , Transforming Growth Factor beta1/antagonists & inhibitors , Adjuvants, Pharmaceutic/pharmacology , Adjuvants, Pharmaceutic/therapeutic use , Nucleotidyltransferases/drug effects , Membrane Proteins/drug effects
4.
Aging (Albany NY) ; 15(24): 15183-15195, 2023 12 27.
Article in English | MEDLINE | ID: mdl-38154100

ABSTRACT

DMC-HA, a novel HDAC inhibitor, has previously demonstrated antiproliferative activity against various cancers, including gliomas. However, the role of DMC-HA in the regulation of EMT and its underlying mechanisms remain unknown. This study aimed to explore the effects of DMC-HA on TGF-ß1-induced EMT in human gliomas and the underlying mechanisms involved. Our results showed that TGF-ß1 induced EMT of U87 and U251 cells, leading to a decrease in epithelial marker ZO-1 and an increase in mesenchymal markers N-cadherin and Vimentin. Moreover, TGF-ß1 treatment resulted in a significant increase in the migratory and invasive abilities of the cells. However, treatment with DMC-HA effectively inhibited the augmented migration and invasion of glioma cells induced by TGF-ß1. Additionally, DMC-HA inhibits TGF-ß1-induced EMT by suppressing canonical Smad pathway and non-canonical TGF-ß/Akt and Erk signalling pathways. These findings suggest that DMC-HA has potential therapeutic implications for gliomas by inhibiting EMT progression.


Subject(s)
Glioma , Transforming Growth Factor beta1 , Humans , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/drug effects , Glioma/drug therapy , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use
5.
Macromol Biosci ; 23(9): e2300184, 2023 09.
Article in English | MEDLINE | ID: mdl-37262314

ABSTRACT

Proper endothelialization and limited collagen deposition on the luminal surface after graft implantation plays a crucial role to prevent the occurrence of stenosis. To achieve these conditions, a biodegradable graft with adequate mechanical properties and the ability to sequentially deliver therapeutic agents isfabricated. In this study, a dual-release system is constructed through coaxial electrospinning by incorporating recombinant human vascular endothelial growth factor (VEGF) and transforming growth factor ß1 (TGF-ß1) inhibitor into silk fibroin (SF) nanofibers to form a bioactive membrane. The functionalized SF membrane as the inner layer of the graft is characterized by the release profile, cell proliferation and protein expression. It presents excellent biocompatibility and biodegradation, facilitating cell attachment, proliferation, and infiltration. The core-shell structure enables rapid VEGF release within 10 days and sustained plasmid delivery for 21 days. A 2.0-mm-diameter vascular graft is fabricated by integrating the SF membrane with decellularized porcine small intestinal submucosa (SIS), aiming to facilitate the integration process under a stable extracellular matrix structure. The bioengineered graft is functionalized with the sequential administration of VEGF and TGF-ß1, and with the reinforced and compatible mechanical properties, thereby offers an orchestrated solution for stenosis with potential for in situ vascular tissue engineering applications.


Subject(s)
Fibroins , Animals , Humans , Constriction, Pathologic , Fibroins/pharmacology , Fibroins/chemistry , Silk/chemistry , Swine , Tissue Engineering , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
6.
Arch Toxicol ; 96(10): 2767-2783, 2022 10.
Article in English | MEDLINE | ID: mdl-35864199

ABSTRACT

Sustained exposure of the lung to various environmental or occupational toxins may eventually lead to pulmonary fibrosis, a devastating disease with no cure. Pulmonary fibrosis is characterized by excessive deposition of extracellular matrix (ECM) proteins such as fibronectin and collagens. The peptidase plasmin degrades the ECM, but protein levels of the plasmin activator inhibitor-1 (PAI-1) are increased in fibrotic lung tissue, thereby dampening plasmin activity. Transforming growth factor-ß1 (TGF-ß1)-induced activation of SMAD transcription factors promotes ECM deposition by enhancing collagen, fibronectin and PAI-1 levels in pulmonary fibroblasts. Hence, counteracting TGF-ß1-induced signaling is a promising approach for the therapy of pulmonary fibrosis. Transient receptor potential cation channel subfamily M Member 7 (TRPM7) supports TGF-ß1-promoted SMAD signaling in T-lymphocytes and the progression of fibrosis in kidney and heart. Thus, we investigated possible effects of TRPM7 on plasmin activity, ECM levels and TGF-ß1 signaling in primary human pulmonary fibroblasts (pHPF). We found that two structurally unrelated TRPM7 blockers enhanced plasmin activity and reduced fibronectin or PAI-1 protein levels in pHPF under basal conditions. Further, TRPM7 blockade strongly inhibited fibronectin and collagen deposition induced by sustained TGF-ß1 stimulation. In line with these data, inhibition of TRPM7 activity diminished TGF-ß1-triggered phosphorylation of SMAD-2, SMAD-3/4-dependent reporter activation and PAI-1 mRNA levels. Overall, we uncover TRPM7 as a novel supporter of TGF-ß1 signaling in pHPF and propose TRPM7 blockers as new candidates to control excessive ECM levels under pathophysiological conditions conducive to pulmonary fibrosis.


Subject(s)
Pulmonary Fibrosis , TRPM Cation Channels , Collagen/antagonists & inhibitors , Collagen/metabolism , Fibrinolysin/metabolism , Fibroblasts , Fibronectins/adverse effects , Fibronectins/antagonists & inhibitors , Fibronectins/metabolism , Fibrosis , Humans , Lung/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Serine-Threonine Kinases , Pulmonary Fibrosis/chemically induced , TRPM Cation Channels/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
7.
Phytomedicine ; 103: 154234, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35689903

ABSTRACT

BACKGROUND: The Modified Shenlingbaizhu Decoction (MSD) utilizes various phytomedicines has been applied to treat colorectal cancer (CRC). Colorectal cancer stem cells (CSCs) have proven to be tightly associated with CRC progression and metastasis. The mechanism of MSD's inhibitory effect on CSCs has not been determined. PURPOSE: To figure out how MSD inhibits the pluripotency of CSCs and impedes the EMT program. METHODS: The ingredients of MSD extracts were characterized by high-performance liquid chromatography (HPLC). BALB/c-nu mice were transplanted into EGFP labeled SW480 CRC cells and the tumor weight and volume were recorded before and after various doses of MSD treatment. The concentration of TGF-ß1 was quantified with an Enzyme-linked immunosorbent assay. To delineate the logical relationship between EMT and CSCs regulated by MSD, TGF-ß/Smad inhibitor and activator were adopted in tumor-bearing mice and diverse CRC cell lines. Cancer stem cell markers were analyzed by flow cytometry. In vitro analysis of cell motility and viability were done using CCK-8, wound healing, and invasion assay. Immunohistochemistry (IHC) and western blotting (WB) were used for detecting protein expression. The collected results were statistically analyzed with GraphPad Prism 8.0. RESULTS: MSD treatment significantly reduced the size of colorectal cancer tumors and lowered the serum content of TGF-ß1 in mice. Importantly, MSD markedly reduced the expression of pluripotent factors and depressed CD133+ stem cells in the tumor tissues. The TGF-ß/Smad inhibitor neutralized the EMT signaling and lowered the pluripotency by dephosphorylation of SMAD2/3. Similarly, MSD attenuated the pluripotency by limiting TGF-ß/Smad signaling-induced EMT in vivo. MSD inhibited colorectal cancer cell proliferation, migration, and invasion. CONCLUSIONS: MSD inhibits the growth of colorectal cancer. It dampens the pluripotency of CSCs by repressing the TGF-ß-induced EMT program.


Subject(s)
Colorectal Neoplasms , Drugs, Chinese Herbal , Neoplastic Stem Cells , Pluripotent Stem Cells , Transforming Growth Factor beta1 , Animals , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/blood , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drugs, Chinese Herbal/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phytotherapy , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/blood
8.
Cell Prolif ; 55(10): e13299, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35762283

ABSTRACT

OBJECTIVES: Ischemia-reperfusion injury (IRI) is a major cause of chronic renal fibrosis. Currently, numerous therapies have shown a minimal effect on the blockade of fibrosis progression. Here, the therapeutic potential of peptide-based TGF-ß1 inhibitor P144 in IRI-induced renal fibrosis and the underlying mechanism were analyzed. MATERIALS AND METHODS: The unilateral ischemia-reperfusion injury with the contralateral nephrectomy model was established, and the P144 was administered intravenously 1d/14d after the onset of IRI. The histopathology and immunofluorescence staining were used to detect renal fibrosis and macrophage infiltration. The in vivo fluorescence imaging was used to measure the bio-distribution of P144. The transwell assays were used to observe the migration of macrophages. RT-qPCR and western blot were used to analyze TGF-ß1 signaling. RESULTS: P144 ameliorated the accumulation of extracellular matrix in the kidney and improved the renal function in the unilateral ischemia-reperfusion injury plus contralateral nephrectomy model. Mechanistically, P144 downregulated the TGF-ß1-Smad3 signaling at both the transcriptional and translational levels and further reduced the TGF-ß1-dependent infiltration of macrophages to the injured kidney. Additionally, P144 blocked the polarization of macrophages to an M2-like phenotype induced by TGF-ß1 in vitro, but showed no effect on their proliferation. CONCLUSIONS: Our study showed that the TGF-ß1 peptide-based inhibitor P144 decreased renal fibrosis through the blockade of the TGF-ß1-Smad3 signaling pathway and the modulation of macrophage polarization, suggesting its potential therapeutic use in IRI-induced renal fibrosis.


Subject(s)
Kidney Diseases , Reperfusion Injury , Transforming Growth Factor beta1 , Fibrosis , Humans , Kidney , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Kidney Diseases/metabolism , Macrophages/metabolism , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
9.
Free Radic Biol Med ; 186: 17-30, 2022 06.
Article in English | MEDLINE | ID: mdl-35513128

ABSTRACT

Oxidative damage and accumulation of extracellular matrix (ECM) components play a crucial role in the adverse outcome of cardiac hypertrophy. Evidence suggests that nuclear factor erythroid-derived factor 2 related factor 2 (Nrf2) can modulate oxidative damage and adverse myocardial remodeling. Daphnetin (Daph) is a coumarin obtained from the plant genus Daphne species that exerts anti-oxidative and anti-inflammatory properties. Herein, we investigated the roles of Daph in transverse aortic constriction (TAC)-induced cardiac hypertrophy and fibrosis in mice. TAC-induced alterations in cardiac hypertrophy markers, histopathological changes, and cardiac function were markedly ameliorated by oral administration of Daph in mice. We found that Daph significantly reduced the reactive oxygen species (ROS) generation, increased the nuclear translocation of Nrf2, and consequently, reinstated the protein levels of NAD(P)H quinone dehydrogenase1 (NQO1), heme oxygenase-1 (HO-1), and other antioxidants in the heart. Besides, Daph significantly inhibited the TAC-induced accumulation of ECM components, including α-smooth muscle actin (α-SMA), collagen I, collagen III, and fibronectin, and interfered with the TGF-ß1/Smad2/3 signaling axis. Further studies revealed that TAC-induced terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) positive nuclei and the protein levels of Bax/Bcl2 ratio and cleaved caspase 3 were substantially decreased by Daph treatment. We further characterized the effect of Daph on angiotensin II (Ang-II)-stimulated H9c2 cardiomyoblast cells and observed that Daph markedly decreased the Ang-II induced increase in cell size, production of ROS, and proteins associated with apoptosis and fibrosis. Mechanistically, Daph alone treatment enhanced the protein levels of Nrf2, NQO1, and HO-1 in H9c2 cells. The inhibition of this axis by Si-Nrf2 transfection abolished the protective effect of Daph in H9c2 cells. Taken together, Daph effectively counteracted the TAC-induced cardiac hypertrophy and fibrosis by improving the Nrf2/HO-1 axis and inhibiting the TGF-ß1/Smad2/3 signaling axis.


Subject(s)
Heme Oxygenase-1 , Membrane Proteins , NF-E2-Related Factor 2 , Smad2 Protein , Smad3 Protein , Transforming Growth Factor beta1 , Umbelliferones , Angiotensin II/metabolism , Animals , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Collagen/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Umbelliferones/pharmacology , Up-Regulation , Ventricular Remodeling/drug effects
10.
BioDrugs ; 36(2): 153-180, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35353346

ABSTRACT

Cancer immunotherapy using monoclonal antibodies targeting immune checkpoints has undoubtedly revolutionized the cancer treatment landscape in the last decade. Immune checkpoint inhibitors can elicit long-lasting, previously unheard-of responses in a number of tumor entities. Yet, even in such tumors as metastatic melanoma and non-small cell-lung cancer, in which immune checkpoint inhibition has become the first-line treatment of choice, only a minority of patients will benefit considerably from these treatments. This has been attributed to a number of factors, including an immune-suppressive tumor microenvironment (TME). Using different modalities to break these barriers is of utmost importance to expand the population of patients that benefit from immune checkpoint inhibition. The multifunctional cytokine transforming growth factor-ß (TGF-ß) has long been recognized as an immune-suppressive factor in the TME. A considerable number of drugs have been developed to target TGF-ß, yet most of these have since been discontinued. The combination of anti-TGF-ß agents with immune checkpoint inhibitors now has the potential to revive this target as a viable immunomodulatory therapeutic approach. Currently, a limited number of small molecular inhibitor and monoclonal antibody candidates that target TGF-ß are in clinical development in combination with the following immune checkpoint inhibitors: SRK 181, an antibody inhibiting the activation of latent TGF-ß1; NIS 793, a monoclonal antibody targeting TGF-ß; and SHR 1701, a fusion protein consisting of an anti-PD-L1 monoclonal antibody fused with the extracellular domain of human TGF-ß receptor II. Several small molecular inhibitors are also in development and are briefly reviewed: LY364947, a pyrazole-based small molecular inhibitor of the serine-threonine kinase activity of TGFßRI; SB-431542, an inhibitor targeting several TGF-ß superfamily Type I activin receptor-like kinases as well as TGF-ß1-induced nuclear Smad3 localization; and galunisertib, an oral small molecular inhibitor of the TGFßRI kinase. One of the most advanced agents in this area is bintrafusp alfa, a bifunctional fusion protein composed of the extracellular domain of TGF-ß receptor II fused to a human IgG1 mAb blocking PD-L1. Bintrafusp alfa is currently in advanced clinical development and as an agent in this space with the most clinical experience, is a focused highlight of this review.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Immunotherapy , Lung Neoplasms , Recombinant Fusion Proteins , Transforming Growth Factor beta , Humans , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/pharmacology , B7-H1 Antigen/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunologic Factors , Lung Neoplasms/drug therapy , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/therapeutic use , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/therapeutic use , Tumor Microenvironment
11.
Life Sci ; 293: 120358, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35092731

ABSTRACT

AIMS: Glioblastoma (GBM) is the most common and aggressive intracranial tumor with poor prognosis. A large majority of clinical chemotherapeutic agents cannot achieve the desired therapeutic effect. Chelerythrine (CHE), a natural component with multitudinous pharmacological functions, has been proven to have outstanding antitumor effects in addition to antibacterial, anti-inflammatory, and hypotensive effects. However, the anti-GBM effect of CHE has not been reported to date. The purpose of this paper is to observe the anti-GBM effect of CHE and further explore the related mechanism. MATERIALS AND METHODS: GBM cell lines (U251 and T98G) and BALB/c nude mice were used in the experiments. Methyl thiazolyl tetrazolium (MTT) and clone formation assays were applied to detect the viability, proliferation and stemness of GBM cells. Flow cytometry was utilized to identify the effect of CHE on GBM apoptosis. Scratch and Transwell experiments reflected the migration and invasion of cells. In vivo, xenograft tumors were implanted subcutaneously in nude mice. The progression of tumors was assessed by ultrasound and magnetic resonance imaging. Finally, western blot, bioinformatics, and immunohistochemistry experiments were used to explore the molecular mechanisms in depth. KEY FINDINGS: In vitro tests showed that CHE inhibited the proliferation, stemness, migration, and invasion of GBM cells and induced apoptosis. In vitro, CHE was observed to restrain the progression of xenograft tumors. We eventually proved that the cytotoxicity of CHE was relevant to the TGFB1-ERK1/2/Smad2/3-Snail/ZEB1 signaling pathway. SIGNIFICANCE: CHE inhibited GBM progression by inhibiting the TGFB1-ERK1/2/Smad2/3-Snail/ZEB1 signaling pathway and is a potential chemotherapeutic drug for GBM.


Subject(s)
Benzophenanthridines/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzophenanthridines/therapeutic use , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Disease Progression , Dose-Response Relationship, Drug , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Smad2 Protein/antagonists & inhibitors , Transforming Growth Factor beta1/antagonists & inhibitors , Xenograft Model Antitumor Assays/methods , Zinc Finger E-box-Binding Homeobox 1/antagonists & inhibitors
12.
Acta Pharmacol Sin ; 43(1): 133-145, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33758354

ABSTRACT

N-n-Butyl haloperidol iodide (F2) is a novel compound that has antiproliferative and antifibrogenic activities. In this study we investigated the therapeutic potential of F2 against liver fibrosis in mice and the underlying mechanisms. Two widely used mouse models of fibrosis was established in mice by injection of either carbon tetrachloride (CCl4) or thioacetamide (TAA). The mice received F2 (0.75, 1.5 or 3 mg·kg-1·d-1, ip) for 4 weeks of fibrosis induction. We showed that F2 administration dose-dependently ameliorated CCl4- or TAA-induced liver fibrosis, evidenced by significant decreases in collagen deposition and c-Jun, TGF-ß receptor II (TGFBR2), α-smooth muscle actin (α-SMA), and collagen I expression in the liver. In transforming growth factor beta 1 (TGF-ß1)-stimulated LX-2 cells (a human hepatic stellate cell line) and primary mouse hepatic stellate cells, treatment with F2 (0.1, 1, 10 µM) concentration-dependently inhibited the expression of α-SMA, and collagen I. In LX-2 cells, F2 inhibited TGF-ß/Smad signaling through reducing the levels of TGFBR2; pretreatment with LY2109761 (TGF-ß signaling inhibitor) or SP600125 (c-Jun signaling inhibitor) markedly inhibited TGF-ß1-induced induction of α-SMA and collagen I. Knockdown of c-Jun decreased TGF-ß signaling genes, including TGFBR2 levels. We revealed that c-Jun was bound to the TGFBR2 promoter, whereas F2 suppressed the binding of c-Jun to the TGFBR2 promoter to restrain TGF-ß signaling and inhibit α-SMA and collagen I upregulation. In conclusion, the therapeutic benefit of F2 against liver fibrosis results from inhibition of c-Jun expression to reduce TGFBR2 and concomitant reduction of the responsiveness of hepatic stellate cells to TGF-ß1. F2 may thus be a potentially new effective pharmacotherapy for human liver fibrosis.


Subject(s)
Haloperidol/analogs & derivatives , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Animals , Carbon Tetrachloride/administration & dosage , Dose-Response Relationship, Drug , Haloperidol/administration & dosage , Haloperidol/pharmacology , Hepatic Stellate Cells/metabolism , Injections, Intraperitoneal , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Structure-Activity Relationship , Thioacetamide/administration & dosage , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
13.
Mol Med Rep ; 25(2)2022 02.
Article in English | MEDLINE | ID: mdl-34913070

ABSTRACT

Pulmonary fibrosis is one of the most important pathological processes associated with paraquat (PQ) poisoning. 5­Aminosalicylic acid (5­ASA) has been shown to be a promising agent against fibrotic diseases. In the present study, the alleviating role of 5­ASA was evaluated in a rat model of pulmonary fibrosis induced by PQ intragastric poisoning (80 mg/kg). Wistar rats were divided into control, PQ, 5­ASA (30 mg/kg daily, 14 days) and PQ + 5­ASA groups. Histological examination revealed congestion, edema and inflammatory cell infiltration in the bronchial and alveolar walls at 3 days after PQ exposure. Alveolar septum thickening with alveolar lumen narrowing was observed at 14 days, while fibroblast proliferation, increase in collagen fiber number and fibrous thickening of the alveolar walls were observed at 28 day. All the aforementioned pulmonary injury changes in the PQ group were attenuated in the PQ + 5­ASA group. Hydroxyproline (HYP) content increased in the lung tissues of the rats at 14 days after PQ treatment and reached a peak at 28 days. Compared with the PQ group, HYP contents of lung tissue decreased at 14 and 28 days after PQ + 5­ASA treatment. Masson's trichrome staining revealed that the increase in the amount of collagen fibers in the lung tissues of rats in the PQ group was inhibited by 5­ASA treatment, further confirming the alleviating effect of 5­ASA on fibrosis. In addition, the results showed that 5­ASA attenuated the upregulation of transforming growth factor­ß1 and phosphorylated­SMAD3, and the reduction of peroxisome proliferator activated receptor γ induced by PQ in lung tissue of rats and human lung fibroblast WI­38 VA13 cells. In conclusion, the results suggested that 5­ASA had an alleviating effect on PQ­induced pulmonary fibrosis, partly by suppressing the activation of the TGF­ß1 signaling pathway.


Subject(s)
Acute Lung Injury/drug therapy , Fibroblasts/drug effects , Lung/drug effects , Mesalamine/administration & dosage , Pulmonary Fibrosis/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Fibroblasts/immunology , Fibroblasts/pathology , Humans , Lung/cytology , Lung/immunology , Lung/pathology , Male , Paraquat/administration & dosage , Paraquat/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Rats , Signal Transduction/drug effects , Signal Transduction/immunology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
14.
Bioorg Chem ; 119: 105573, 2022 02.
Article in English | MEDLINE | ID: mdl-34952245

ABSTRACT

Tetrodecadazinone (1), a novel tetrodecamycin-pyridazinone hybrid possessing a new 1,2-dimethyl-1-(2-methylnonyl)decahydronaphthalene skeleton, and 4-hydroxydihydrotetrodecamycin (2) were separated from a culture of Streptomyces sp. HU051, together with a known compound, dihydrotetrodecamycin (3). Diverse spectroscopic approaches were applied to assign the structures of 1-3, and the structure of 1 was further confirmed by single crystal X-ray diffraction analysis. Compound 1 is the first example of a pyridazinone-containing natural product. Biosynthetically, 1 is proposed to be derived from a Michael addition reaction of a PKS-derived tetrodecamycin and a piperazic-acid-derived pyridazinone. Biological evaluation revealed 1 could reduce the expressions of extracellular matrix proteins (fibronectin and collagen I) and α-smooth muscle actin (α-SMA) in transforming growth factor-ß (TGF-ß1)-activated LX-2 cells. Preliminary mechanism study showed 1 exerted its anti-liver fibrosis effect by regulating TGF-ß1/Smad2/3 signaling pathway.


Subject(s)
Anti-Bacterial Agents/pharmacology , Liver Cirrhosis/drug therapy , Streptomyces/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/microbiology , Microbial Sensitivity Tests , Molecular Conformation , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/metabolism , Structure-Activity Relationship , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism
15.
Am J Chin Med ; 49(8): 1965-1999, 2021.
Article in English | MEDLINE | ID: mdl-34961416

ABSTRACT

Pulmonary fibrosis (PF) is a chronic and irreversible interstitial lung disease that even threatens the lives of some patients infected with COVID-19. PF is a multicellular pathological process, including the initial injuries of epithelial cells, recruitment of inflammatory cells, epithelial-mesenchymal transition, activation and differentiation of fibroblasts, etc. TGF-[Formula: see text]1 acts as a key effect factor that participates in these cellular processes of PF. Recently, much attention was paid to inhibiting TGF-[Formula: see text]1 mediated cell processes in the treatment of PF with Chinese herbal medicines (CHM), an important part of traditional Chinese medicine. Here, this review first summarized the effects of TGF-[Formula: see text]1 in different cellular processes of PF. Then, this review summarized the recent research on CHM (compounds, multi-components, single medicines and prescriptions) to directly and/or indirectly inhibit TGF-[Formula: see text]1 signaling (TLRs, PPARs, micrRNA, etc.) in PF. Most of the research focused on CHM natural compounds, including but not limited to alkaloids, flavonoids, phenols and terpenes. After review, the research perspectives of CHM on TGF-[Formula: see text]1 inhibition in PF were further discussed. This review hopes that revealing the inhibiting effects of CHM on TGF-[Formula: see text]1-mediated cellular processes of PF can promote CHM to be better understood and utilized, thus transforming the therapeutic activities of CHM into practice.


Subject(s)
Cell Physiological Phenomena/drug effects , Drugs, Chinese Herbal/therapeutic use , Pulmonary Fibrosis/drug therapy , Signal Transduction/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , COVID-19/complications , COVID-19/metabolism , COVID-19/virology , Humans , Medicine, Chinese Traditional/methods , Phytotherapy/methods , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/metabolism , SARS-CoV-2/physiology , Transforming Growth Factor beta1/metabolism
16.
Eur J Pharmacol ; 912: 174550, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34610279

ABSTRACT

BACKGROUND: The interaction between tumor cells and tumor microenvironment is a necessary condition for promoting the metastasis of malignant tumors. METHODS: Two different transwell culture systems were interfered with by recombinant factor placental growth factor (re-PIGF) and the re-PIGF + transforming growth factor-ß1 (TGF-ß1)-neutralizing antibody (anti-TGF-ß1). We performed immunofluorescence, flow cytometry and enzyme linked immunosorbent assay (ELISA) to analyze the expression of PIGF, fms-like tyrosine kinase-1 (Flt-1), macrophage marker F4/80 +, macrophage M2 marker CD163+ and TGF-ß1 in vitro. Meanwhile, cell viability assay and optical microscope assay were conducted to explore the cell viability and vascularization ability of human umbilical vein endothelial cells (HUVECs). RESULTS: Re-PIGF increased the expression of PIGF in A549 cells and the expression of Flt-1 in BM-Mac cells, and significantly enhanced the ability of bone marrow-derived macrophages (BM-Mac) to transform into macrophages. At the same time, re-PIGF increased the expression of cytokine TGF-ß1 in A549 cells/BM-Mac transwell culture system. On the contrary, re-PIGF + anti-TGF-ß1 inhibited the expression of Flt-1 in BM-Mac cells and inhibited the ability of BM-Mac cells to transform into macrophages. Finally, re-PIGF + anti-TGF-ß1 reduced the cell viability and angiogenesis of HUVECs. CONCLUSION: The surface molecule PIGF of lung cancer cells could bind to the receptor Flt-1 on the surface of macrophages, thereby increasing the production of TGF-ß1, and ultimately promoting the formation of angiogenesis in lung cancer.


Subject(s)
Lung Neoplasms/blood supply , Lung Neoplasms/immunology , Neovascularization, Pathologic/metabolism , Placenta Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Tumor-Associated Macrophages/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , A549 Cells , Angiogenesis Inducing Agents/pharmacology , Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Survival/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Lung Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Recombinant Proteins/pharmacology , Transforming Growth Factor beta1/antagonists & inhibitors , Tumor-Associated Macrophages/drug effects , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
17.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638597

ABSTRACT

Cyclosporine A (CsA) is a nephrotoxicant that causes fibrosis via induction of epithelial-mesenchymal transition (EMT). The flavonoid chrysin has been reported to have anti-fibrotic activity and inhibit signaling pathways that are activated during EMT. This study investigated the nephroprotective role of chrysin in the prevention of CsA-induced renal fibrosis and elucidated a mechanism of inhibition against CsA-induced EMT in proximal tubule cells. Treatment with chrysin prevented CsA-induced renal dysfunction in Sprague Dawley rats measured by blood urea nitrogen (BUN), serum creatinine and creatinine clearance. Chrysin inhibited CsA-induced tubulointerstitial fibrosis, characterized by reduced tubular damage and collagen deposition. In vitro, chrysin significantly inhibited EMT in LLC-PK1 cells, evidenced by inhibition of cell migration, decreased collagen expression, reduced presence of mesenchymal markers and elevated epithelial junction proteins. Furthermore, chrysin co-treatment diminished CsA-induced TGF-ß1 signaling pathways, decreasing Smad 3 phosphorylation which lead to a subsequent reduction in Snail expression. Chrysin also inhibited activation of the Akt/ GSK-3ß pathway. Inhibition of both pathways diminished the cytosolic accumulation of ß-catenin, a known trigger for EMT. In conclusion, flavonoids such as chrysin offer protection against CsA-induced renal dysfunction and interstitial fibrosis. Chrysin was shown to inhibit CsA-induced TGF-ß1-dependent EMT in proximal tubule cells by modulation of Smad-dependent and independent signaling pathways.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Fibrosis/drug therapy , Flavonoids/pharmacology , Kidney Diseases/drug therapy , Kidney Diseases/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Cell Movement/drug effects , Collagen/metabolism , Cyclosporine/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibrosis/chemically induced , Fibrosis/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad3 Protein/metabolism
18.
Mol Pharm ; 18(11): 4140-4147, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34657437

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease with poor prognosis. Evidence has shown that vimentin is a key regulator of lung fibrogenesis. 99mTc-labeled N-acetylglucosamine-polyethyleneimine (NAG-PEI), a vimentin-targeting radiotracer, was used for the early diagnosis of IPF, and NAG-PEI was also used as a therapeutic small interfering RNA (siRNA) delivery vector for the treatment of IPF in this study. Single-photon emission-computed tomography (SPECT) imaging of bleomycin (BM)- and silica-induced IPF mice with 99mTc-labeled NAG-PEI was performed to visualize pulmonary fibrosis and monitor the treatment efficiency of siRNA-loaded NAG-PEI, lipopolysaccharide (LPS, a tolerogenic adjuvant), or zymosan (ZYM, an immunostimulant). The lung uptakes of 99mTc-NAG-PEI in the BM- and silica-induced IPF mice were clearly and directly correlated with IPF progression. The lung uptake of 99mTc-NAG-PEI in the NAG-PEI/TGF-ß1-siRNA treatment group or LPS treatment group was evidently lower than that in the control group, while the lung uptake of 99mTc-NAG-PEI was significantly higher in the ZYM treatment group compared to that in the control group. These results demonstrate that NAG-PEI is a potent MicroSPECT imaging-guided theranostic platform for IPF diagnosis and therapy.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , RNA, Small Interfering/administration & dosage , Radiopharmaceuticals/administration & dosage , Transforming Growth Factor beta1/antagonists & inhibitors , Vimentin/antagonists & inhibitors , Acetylglucosamine/administration & dosage , Acetylglucosamine/chemistry , Animals , Biodiversity , Bleomycin/administration & dosage , Bleomycin/toxicity , Disease Models, Animal , Female , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/pathology , Lung/diagnostic imaging , Lung/drug effects , Lung/pathology , Mice , Polyethyleneimine/administration & dosage , Polyethyleneimine/chemistry , RNA, Small Interfering/genetics , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Silicon Dioxide/administration & dosage , Silicon Dioxide/toxicity , Technetium , Tomography, Emission-Computed, Single-Photon , Transforming Growth Factor beta1/metabolism , Vimentin/metabolism
19.
Physiol Res ; 70(5): 815-820, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34505520

ABSTRACT

Transforming growth factor beta 1 (TGF-beta1) is a pro-fibrotic cytokine with a key role in wound repair and regeneration, including induction of fibroblast-to-myofibroblast transition. Genistein is a naturally occurring selective estrogen receptor modulator with promising anti-fibrotic properties. In the present study we aimed to investigate whether genistein modulates TGF-beta1 (canonical and non-canonical) signaling in normal dermal fibroblasts at the protein level (Western blot and immunofluorescence). We demonstrated that TGF-beta1 induces the myofibroblast-like phenotype in the studied fibroblast signaling via canonical (SMAD) and non-canonical (AKT, ERK1/2, ROCK) pathways. Genistein induced only ERK1/2 expression, whereas the combination of TGF-beta1 and genistein attenuated the ERK1/2 and ROCK signaling. Of note, the other studied pathways remained almost unaffected. From this point of view, genistein does not impair conversion of normal fibroblasts to myofibroblast-like cells.


Subject(s)
Fibroblasts/drug effects , Genistein/pharmacology , Phytoestrogens/pharmacology , Transforming Growth Factor beta1/metabolism , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Humans , Primary Cell Culture , Signal Transduction/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors
20.
Pharmacology ; 106(11-12): 667-681, 2021.
Article in English | MEDLINE | ID: mdl-34518478

ABSTRACT

INTRODUCTION: Silent information regulator 1 (SIRT1) has been extensively investigated in the cardiovascular system and has been shown to play a pivotal role in mediating cell death/survival, energy production, and oxidative stress. However, the functional role of SIRT1 in pressure overload-induced cardiac hypertrophy and dysfunction remains unclear. Resveratrol (Rsv), a widely used activator of SIRT1, has been reported to protect against cardiovascular disease. We here examine whether activation of SIRT1 by Rsv attenuate pressure overload-induced cardiac hypertrophy and to identify the underlying molecular mechanisms. METHODS: In vivo, rat model of pressure overload-induced myocardial hypertrophy was established by abdominal aorta constriction (AAC) procedure. In vitro, Angiotensin II (Ang II) was applied to induce hypertrophy in cultured neonatal rat cardiomyocytes (NCMs). Hemodynamics and histological analyses of the heart were evaluated. The expression of SIRT1, transforming growth factor-ß1 (TGF-ß1)/phosphorylated (p)-small mother against decapentaplegic (Smad)3 and hypertrophic markers were determined by immunofluorescence, real-time PCR, and Western blotting techniques. RESULTS: In the current study, Rsv treatment improved left ventricular function and reduced left ventricular hypertrophy and cardiac fibrosis significantly in the pressure overload rats. The expression of SIRT1 was significantly reduced, while the expression of TGF-ß1/p-Smad3 was significantly enhanced in AAC afflicted rat heart. Strikingly, treatment with Rsv restored the expressions of SIRT1 and TGF-ß1/p-Smad3 under AAC influence. However, SIRT1 inhibitor Sirtinol (Snl) markedly prevented the effects of Rsv, which suggest that SIRT1 signaling pathway was involved in the cardiac protective effect of Rsv. In vitro studies performed in Ang II-induced hypertrophy in NCMs confirmed the cardiac protective effect of Rsv. Furthermore, the study presented that SIRT1 negatively correlated with the cardiac hypertrophy, cardiac fibrosis, and the TGF-ß1/p-Smad3 expression. CONCLUSIONS: Taken together, these results indicated that activation of SIRT1 by Rsv attenuates cardiac hypertrophy, cardiac fibrosis, and improves cardiac function possibly via regulation of the TGF-ß1/p-Smad3 signaling pathway. Our study may provide a potential therapeutic strategy for cardiac hypertrophy.


Subject(s)
Cardiomegaly/pathology , Resveratrol/pharmacology , Sirtuin 1/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Disease Models, Animal , Fibrosis/pathology , Hemodynamics/drug effects , Hypertrophy, Left Ventricular/pathology , Male , Muscle Cells/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , Smad3 Protein/drug effects , Ventricular Function/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...