Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Cardiothorac Surg ; 19(1): 208, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38616256

ABSTRACT

BACKGROUND: Cardiac fibroblasts (CFs) are activated after initial injury, and then differentiate into myofibroblasts (MFs), which play a pivotal role as the primary mediator cells in pathological remodeling. Sodium butyrate (NaB), being a metabolite of gut microbiota, exhibits anti-inflammatory property in local therapies on sites other than the intestine. Thus, this study aimed to probe the mechanism by which NaB regulates CFs transdifferentiation through the NLRP3/Caspase-1 pyroptosis pathway. METHODS: CFs were cultured in vitro and induced into MFs by TGFß1. CFs were identified by immunofluorescence labelling technique of vimentin and α-SMA, followed by treatment with NaB or NLRP3 inflammasome inhibitor (CY-09) and its activator [nigericin sodium salt (NSS)]. The expression levels of α-SMA, GSDMD-N/NLRP3/cleaved Caspase-1 proteins, and inflammatory factors IL-1ß/IL-18/IL-6/IL-10 were determined using immunofluorescence, Western blot and ELISA. Cell proliferation and migration were evaluated using the CCK-8 assay and the cell scratch test, respectively. RESULTS: Following the induction of TGFß1, CFs exhibited increased expression levels of α-SMA proteins and IL-6/IL-10, as well as cell proliferative and migratory abilities. TGFß1 induced CFs to differentiate into MFs, while NaB inhibited this differentiation. NaB inactivated the NLRP3/Caspase-1 pyroptosis pathway. CY-09 demonstrated inhibitory effects on the NLRP3/Caspase-1 pyroptosis pathway, leading to a reduction in TGFß1-induced CFs transdifferentiation. NSS activated the NLRP3/Caspase-1 pyroptosis pathway, and thus partially counteracting the inhibitory effect of intestinal microbiota metabolite NaB on CFs transdifferentiation. CONCLUSION: NaB, a metabolite of the gut microbiota, inhibited the activation of the NLRP3/Caspase-1 pyroptosis pathway in TGFß1-induced CFs, repressed the transdifferentiation of CFs into MFs.


Subject(s)
Gastrointestinal Microbiome , Humans , Caspase 1 , NLR Family, Pyrin Domain-Containing 3 Protein , Butyric Acid , Interleukin-10 , Cell Transdifferentiation , Interleukin-6 , Pyroptosis , Fibroblasts
2.
Drug Des Devel Ther ; 16: 2517-2527, 2022.
Article in English | MEDLINE | ID: mdl-35946039

ABSTRACT

Purpose: Endothelial-to-mesenchymal transition (EndMT) is an important mechanism underlying cardiac fibrosis. The anti-ischemic drug trimetazidine (TMZ) is reportedly useful in ventricular remodeling and associated with NADPH oxidase (NOX) 2. This study aimed to investigate the possible effect of TMZ on cardiac fibrosis exerted via the inhibition of NOX2-mediated EndMT. Methods: A cardiac fibrosis model was established in Sprague-Dawley rats through a subcutaneous injection of isoproterenol (ISO, 5 mg/kg/d). Echocardiographic parameters, myocardial fibrosis, NOX2 expression and EndMT were assessed. An in vitro model of EndMT was developed using human umbilical vein endothelial cells (HUVECs) via treatment with transforming growth factor-ß (TGF-ß) at 10 ng/mL for 24 h. HUVECs were administrated with TMZ or TMZ and lentivirus, the expression of EndMT and related proteins was observed by wound healing assay, immunoblotting, and immunofluorescence. Results: Rats injected with ISO exhibited severe interstitial cardiac fibrosis and perivascular fibrosis, decreased left ventricular ejection fraction, and increased NOX activity. TMZ treatment mitigated cardiac fibrosis, ameliorated left ventricular dysfunction, and reduced NOX activity. In addition, TMZ effectively inhibited EndMT in ISO-treated rat hearts and TGF-ß-treated HUVECs, as manifested by increased CD31 expression, decreased α-SMA expression, and suppressed cell migration. Compared with the control group, the expression of NOX2, nuclear factor-κB (NF-κB), and Snail was increased in vivo and in vitro but decreased with TMZ treatment. Furthermore, the overexpression of NOX2 by lentivirus abolished the protective effects of TMZ on TGF-ß-induced EndMT. Conclusion: TMZ may ameliorate EndMT and ISO-induced cardiac fibrosis through the NOX2/NF-κB/Snail pathway. The findings of the study may provide new insights into the potential role of TMZ in the pathophysiology of cardiac fibrosis.


Subject(s)
Cardiomyopathies , Trimetazidine , Animals , Cardiomyopathies/metabolism , Epithelial-Mesenchymal Transition , Fibrosis , Human Umbilical Vein Endothelial Cells , Humans , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Stroke Volume , Transforming Growth Factor beta/metabolism , Trimetazidine/metabolism , Trimetazidine/pharmacology , Ventricular Function, Left
4.
Acta Pharmacol Sin ; 42(5): 814-823, 2021 May.
Article in English | MEDLINE | ID: mdl-32855532

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most widespread type of non-Hodgkin lymphoma (NHL). As the most aggressive form of the DLBCL, the activated B-cell-like (ABC) subtype is often resistant to standard chemotherapies. Bruton's tyrosine kinase (BTK) inhibitor ibrutinib provides a potential therapeutic approach for the DLBCL but fails to improve the outcome in the phase III trial. In the current study, we investigated the molecular mechanisms underlying ibrutinib resistance and explored new combination therapy with ibrutinib. We generated an ibrutinib-resistant ABC-DLBCL cell line (OCI-ly10-IR) through continuous exposure to ibrutinib. Transcriptome analysis of the parental and ibrutinib-resistant cell lines revealed that the ibrutinib-resistant cells had significantly lower expression of the unfolded protein response (UPR) marker genes. Overexpression of one UPR branch-XBP1s greatly potentiated ibrutinib-induced apoptosis in both sensitive and resistant cells. The UPR inhibitor tauroursodeoxycholic acid (TUDCA) partially reduced the apoptotic rate induced by the ibrutinib in sensitive cells. The UPR activator 2-deoxy-D-glucose (2-DG) in combination with the ibrutinib triggered even greater cell growth inhibition, apoptosis, and stronger calcium (Ca2+) flux inhibition than either of the agents alone. A combination treatment of ibrutinib (15 mg·kg-1·d-1, po.) and 2-DG (500 mg/kg, po, b.i.d.) synergistically retarded tumor growth in NOD/SCID mice bearing OCI-ly10-IR xenograft. In addition, ibrutinib induced the UPR in the sensitive cell lines but not in the resistant cell lines of the DLBCL. There was also a combined synergistic effect in the primary resistant DLBCL cell lines. Overall, our results suggest that targeting the UPR could be a potential combination strategy to overcome ibrutinib resistance in the DLBCL.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Lymphoma, Large B-Cell, Diffuse/drug therapy , Piperidines/therapeutic use , Unfolded Protein Response/drug effects , Adenine/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxyglucose/therapeutic use , Drug Resistance, Neoplasm/physiology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/physiopathology , Mice, Inbred NOD , Mice, SCID , Unfolded Protein Response/physiology , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Xenograft Model Antitumor Assays
5.
Aging (Albany NY) ; 12(21): 22112-22121, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33186920

ABSTRACT

Mesenchymal-to-endothelial transition (MEndT) is one of the mechanisms that influences cardiac fibrosis, which is a key process in cardiac remodeling. It has been reported that autophagy inhibits endothelial cell transition. However, whether autophagy could modulate MEndT in cardiac fibrosis has not yet been investigated. Here, we discussed the association between autophagy and MEndT and its possible mechanism. In this study, we induced endothelial-to-mesenchymal transition using transforming growth factor-ß to generate mesenchymal cells and fibroblasts in wild-type human umbilical vein endothelial cells and cells with p53 knockout or overexpression. Then, autophagy was induced by Earle's balanced salt solution (EBSS) and was inhibited by bafilomycin A1 or lentivirus-ATG5-shRNA. The expression levels of MEndT and the autophagy markers CD31, VE-Cadherin, Vimentin, α-SMA, LC3, p62 and p53 were examined. We found that activation of autophagy could promote MEndT and increase cytoplasmic and total expression of p53, that but nuclear p53 expression was decreased, and that inhibition of autophagy activation could reverse the effect of EBSS. Moreover, after knockout of nuclear p53, autophagy promoted MEndT, while autophagy inhibited MEndT in p53 overexpressing cells. Our results demonstrate that autophagy modulate MEndT by nuclear p53 provide a new strategy for the treatment of fibrosis diseases.


Subject(s)
Autophagy , Epithelial-Mesenchymal Transition , Human Umbilical Vein Endothelial Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Cells, Cultured , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/pathology , Humans , Macrolides/pharmacology , Signal Transduction , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Protein p53/genetics
6.
Diabetes ; 68(12): 2197-2209, 2019 12.
Article in English | MEDLINE | ID: mdl-31471292

ABSTRACT

Uncoupling of mitochondrial respiration by chemical uncouplers has proven effective in ameliorating obesity, insulin resistance, and hyperglycemia. However, development of uncoupler-based therapy remains challenging due to its potentially lethal adverse effects. Here, we identify pyruvate dehydrogenase (PDH) as a key modifier of the toxicity profile of 2, 4-dinitrophenol (DNP), a prototypical mitochondrial uncoupler. PDH activation by dichloroacetic acid (DCA) protects mice from DNP-induced hyperlactacidemia, hyperthermia, and death while preserving the ability of DNP to promote fuel oxidation and improve insulin sensitivity in mice. Mechanistically, PDH activation switches on mitochondrial glucose oxidation to accommodate increased glycolytic flux, leading to reduced lactate secretion during uncoupler treatments. We devised a chemical screening strategy and discovered compound 6j as a dual-action compound that simultaneously activates PDH and uncouples mitochondrial respiration. Compound 6j exhibits an excellent efficacy and safety profile in restoring glucose homeostasis in diabetic mice. This work establishes a new principle to safely harness the power of chemical uncouplers for the treatment of metabolic disease.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , Animals , Glucose , Mice , Oxidoreductases , Pyruvate Dehydrogenase Complex , Pyruvates
7.
Eur J Med Chem ; 180: 398-416, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31325786

ABSTRACT

In this work, aiming at finding a novel, potent, and selective sodium-dependent glucose cotransporter 2 (SGLT2) inhibitor with good pharmacokinetic profiles for the treatment of diabetes, we focus on modifying the sugar moiety of SGLT2 inhibitors, which dominates the binding with glucose binding site of hSGLT, via removing the C-6 hydroxy group to adjust the physicochemical properties and target-recognition manners of SGLT2 inhibitors. In addition, tofogliflozin containing a special O-spiroketal C-arylglucoside scaffold, displayed good efficacy and bioavailability both in animals and in humans. Therefore, a series of 6-deoxy O-spiroketal C-arylglucosides as novel SGLT2 inhibitors were designed, synthesized, and evaluated in this work. The structure-activity relationship (SAR) research on this novel series and a comprehensive in vitro and in vivo biological evaluation afforded compound 39 with high in vitro hSGLT2 inhibitory activity (IC50 = 4.5 nM), good pharmacokinetic profiles, and more remarkable efficacy in C57BL/6J mice and Sprague-Dawley rats than marketed drug tofogliflozin.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drug Design , Sodium-Glucose Transporter 2 Inhibitors/chemical synthesis , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Animals , Benzhydryl Compounds/chemical synthesis , Benzhydryl Compounds/chemistry , Benzhydryl Compounds/pharmacology , Dose-Response Relationship, Drug , Glucosides/chemical synthesis , Glucosides/chemistry , Glucosides/pharmacology , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 2 Inhibitors/chemistry , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Structure-Activity Relationship
8.
J Cell Mol Med ; 23(9): 6355-6367, 2019 09.
Article in English | MEDLINE | ID: mdl-31297954

ABSTRACT

Long non-coding RNAs (lncRNAs) have emerged as potent regulators of cardiac disease; however, the role of lncRNA in cardiac fibrosis remains partially understood. In this study, we identified a cardiac endothelial-enriched lncRNA-lnc000908, which was markedly up-regulated in rats with cardiac fibrosis. In addition, the expression of prostaglandin E2 receptor 4 (EP4) was decreased in cardiac fibrosis. In vivo lnc000908 silencing by lentivirus increased the EP4 level, decreased endothelial-mesenchymal transition (EndMT) and improved cardiac fibrosis and cardiac function. Consistently, the lnc000908 knockdown also up-regulated EP4 and suppressed transforming growth factor-beta (TGF-ß)-induced EndMT in cardiac microvascular endothelial cells. In contrast, the lnc000908 overexpression by lentivirus decreased the EP4 level and induced EndMT. Of note, these pro- or anti-EndMT effects were reversed by the EP4 overexpression or the EP4 antagonist AH-23848, respectively. This study demonstrates that lnc000908 is a novel regulator of cardiac fibrosis by modulating the EP4 expression and EndMT.


Subject(s)
Endothelium, Vascular/metabolism , Epithelial-Mesenchymal Transition/physiology , RNA, Long Noncoding/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Ventricular Remodeling/physiology , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Fibrosis/metabolism , Fibrosis/pathology , Heart/physiology , Male , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/metabolism
9.
Life Sci ; 224: 177-186, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30658104

ABSTRACT

AIMS: The mechanisms underlying cardiorenal syndromes are complex and not fully understood; Fibrosis seems to be a primary driver of the diseases' pathophysiology. Spironolactone can reduce cardiac or renal fibrosis by inhibiting endothelial-mesenchymal transition (EndMT). Spironolactone protection may rely on activation of adenosine receptors, but the role of the adenosine A2A receptor (A2AR) is unclear. We hypothesize that spironolactone may modulate A2AR to suppress EndMT and reduce cardiorenal remodeling. MAIN METHODS: A model of renal injury followed by heart failure was established by subcutaneous administration of isoprenaline (Iso) to rats. Assessment of cardiac and renal function, fibrosis, EndMT markers, adenosine and A2AR expression was performed. TGF-ß was used to induce EndMT in primary human umbilical vein endothelial cells (HUVECs). Rats or cells were divided into four groups: those that treated with spironolactone alone or in combination with A2AR antagonist ZM241385 or neither, and compared to normal controls. KEY FINDINGS: Isoprenaline-treated rats exhibited cardiac and renal fibrosis, impaired cardiac and renal function, enhanced EndMT, and lower A2AR expression. Spironolactone significantly up-regulated A2AR expression and inhibited EndMT in vivo and in vitro. Moreover, spironolactone improved cardiorenal remodeling and reduced dysfunction. These changes were exacerbated by administration of ZM241385. Together, these findings show that spironolactone up-regulated A2AR to reduce EndMT and ameliorate cardiorenal fibrosis. SIGNIFICANCE: The anti-fibrotic effects of spironolactone may partly depend on the up-regulation of A2AR, and that A2AR might be a potential therapeutic target for the treatment of cardiorenal syndrome.


Subject(s)
Cardio-Renal Syndrome/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Fibrosis/drug therapy , Mineralocorticoid Receptor Antagonists/pharmacology , Receptor, Adenosine A2A/metabolism , Spironolactone/pharmacology , Animals , Cardio-Renal Syndrome/metabolism , Cardio-Renal Syndrome/pathology , Cells, Cultured , Fibrosis/metabolism , Fibrosis/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Male , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics
10.
Eur J Med Chem ; 86: 242-56, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25164763

ABSTRACT

Based on the previous work in our group and the principle of computer-aided drug design, a series of novel ß-amino pyrrole-2-carbonitrile derivatives was designed and synthesized. Compounds 8l and 9l were efficacious and selective DPP4 inhibitors resulting in decreased blood glucose in vivo. Compound 8l had moderate DPP4 inhibitory activity (IC50 = 0.05 µM) and good oral bioavailability (F = 53.2%). Compound 9l showed excellent DPP4 inhibitory activity (IC50 = 0.01 µM), good selectivity (selective ratio: DPP8/DPP4 = 898.00; DPP9/DPP4 = 566.00) against related peptidases, and good efficacy in an oral glucose tolerance tests in ICR mice and moderate PK profiles (F = 22.8%, t1/2 = 2.74 h). Moreover, compound 9l did not block hERG channel and exhibited no inhibition of liver metabolic enzymes such as CYP2C9.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Drug Design , Nitriles/pharmacology , Animals , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Mice , Mice, Inbred ICR , Mice, Obese , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry , Structure-Activity Relationship
11.
PLoS One ; 8(8): e72092, 2013.
Article in English | MEDLINE | ID: mdl-23977216

ABSTRACT

AMP-activated protein kinase (AMPK) is an energy sensor of metabolism that is an attractive therapeutic target for type 2 diabetes mellitus and metabolic syndrome. Using a homogeneous scintillation proximity assay (SPA), we identified a new small-molecule AMPK activator, ZLN024, which allosterically stimulated active AMPK heterotrimers and the inactive α1 subunit truncations α1 (1-394) and α1 (1-335) but not α1 (1-312). AMPK activation by ZLN024 requires the pre-phosphorylation of Thr-172 by at least one upstream kinase and protects AMPK Thr-172 against dephosphorylation by PP2Cα. ZLN024 activated AMPK in L6 myotubes and stimulated glucose uptake and fatty acid oxidation without increasing the ADP/ATP ratio. ZLN024 also activated AMPK in primary hepatocytes, decreased fatty acid synthesis and glucose output. Treatment of db/db mice with 15 mg/kg/day ZLN024 improved glucose tolerance; liver tissue weight, triacylglycerol and the total cholesterol content were decreased. The hepatic transcriptional level of G6Pase, FAS and mtGPAT were reduced. The transcription of genes involved in fatty acid oxidation and the mitochondrial biogenesis of muscle tissue were elevated. The ACC phosphorylation was increased in muscle and liver. This study provides a novel allosteric AMPK activator for functional study in vitro and in vivo and demonstrates that AMPK allosteric activators could be a promising therapeutic approach for type 2 diabetes mellitus and metabolic syndrome.


Subject(s)
Adenylate Kinase/metabolism , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activators/pharmacology , Hypoglycemic Agents/pharmacology , Pyrimidines/pharmacology , Adenosine Triphosphate/metabolism , Allosteric Regulation , Animals , Blood Glucose , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Enzyme Activators/therapeutic use , Fatty Acids/metabolism , Glucose/metabolism , HeLa Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/therapeutic use , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Obese , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Primary Cell Culture , Protein Phosphatase 2C , Protein Processing, Post-Translational , Pyrimidines/therapeutic use , Rats
12.
Diabetes ; 62(4): 1297-307, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23250358

ABSTRACT

Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) has been shown to influence energy metabolism. Hence, we explored a strategy to target PGC-1α expression to treat metabolic syndromes. We developed a high-throughput screening assay that uses the human PGC-1α promoter to drive expression of luciferase. The effects of lead compound stimulation on PGC-1α expression in muscle cells and hepatocytes were investigated in vitro and in vivo. A novel small molecule, ZLN005, led to changes in PGC-1α mRNA levels, glucose uptake, and fatty acid oxidation in L6 myotubes. Activation of AMP-activated protein kinase was involved in the induction of PGC-1α expression. In diabetic db/db mice, chronic administration of ZLN005 increased PGC-1α and downstream gene transcription in skeletal muscle, whereas hepatic PGC-1α and gluconeogenesis genes were reduced. ZLN005 increased fat oxidation and improved the glucose tolerance, pyruvate tolerance, and insulin sensitivity of diabetic db/db mice. Hyperglycemia and dyslipidemia also were ameliorated after treatment with ZLN005. Our results demonstrated that a novel small molecule selectively elevated the expression of PGC-1α in myotubes and skeletal muscle and exerted promising therapeutic effects for treating type 2 diabetes.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Gene Expression Regulation/drug effects , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Trans-Activators/metabolism , Adipose Tissue/metabolism , Animals , Dyslipidemias , Gluconeogenesis/drug effects , Gluconeogenesis/physiology , Glucose Intolerance , HEK293 Cells , Humans , Hyperglycemia , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred NOD , Molecular Structure , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Random Allocation , Trans-Activators/genetics , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...