Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2983, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38582860

ABSTRACT

Akkermansia muciniphila has received great attention because of its beneficial roles in gut health by regulating gut immunity, promoting intestinal epithelial development, and improving barrier integrity. However, A. muciniphila-derived functional molecules regulating gut health are not well understood. Microbiome-secreted proteins act as key arbitrators of host-microbiome crosstalk through interactions with host cells in the gut and are important for understanding host-microbiome relationships. Herein, we report the biological function of Amuc_1409, a previously uncharacterised A. muciniphila-secreted protein. Amuc_1409 increased intestinal stem cell (ISC) proliferation and regeneration in ex vivo intestinal organoids and in vivo models of radiation- or chemotherapeutic drug-induced intestinal injury and natural aging with male mice. Mechanistically, Amuc_1409 promoted E-cadherin/ß-catenin complex dissociation via interaction with E-cadherin, resulting in the activation of Wnt/ß-catenin signaling. Our results demonstrate that Amuc_1409 plays a crucial role in intestinal homeostasis by regulating ISC activity in an E-cadherin-dependent manner and is a promising biomolecule for improving and maintaining gut health.


Subject(s)
Verrucomicrobia , beta Catenin , Male , Mice , Animals , beta Catenin/metabolism , Verrucomicrobia/metabolism , Intestines , Cadherins/metabolism , Akkermansia
2.
Life Sci ; 339: 122413, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38219919

ABSTRACT

AIMS: The gut microbiota is increasingly recognised as a pivotal regulator of immune system homeostasis and brain health. Recent research has implicated the gut microbiota in age-related cognitive impairment and dementia. Agathobaculum butyriciproducens SR79 T (SR79), which was identified in the human gut, has been reported to be beneficial in addressing cognitive deficits and pathophysiologies in a mouse model of Alzheimer's disease. However, it remains unknown whether SR79 affects age-dependent cognitive impairment. MAIN METHOD: To explore the effects of SR79 on cognitive function during ageing, we administered SR79 to aged mice. Ageing-associated behavioural alterations were examined using the open field test (OFT), tail suspension test (TST), novel object recognition test (NORT), Y-maze alternation test (Y-maze), and Morris water maze test (MWM). We investigated the mechanisms of action in the gut and brain using molecular and histological analyses. KEY FINDINGS: Administration of SR79 improved age-related cognitive impairment without altering general locomotor activity or depressive behaviour in aged mice. Furthermore, SR79 increased mature dendritic spines in the pyramidal cells of layer III and phosphorylation of CaMKIIα in the cortex of aged mice. Age-related activation of astrocytes in the cortex of layers III-V of the aged brain was reduced following SR79 administration. Additionally, SR79 markedly increased IL-10 production and Foxp3 and Muc2 mRNA expression in the colons of aged mice. SIGNIFICANCE: These findings suggest that treatment with SR79 may be a beneficial microbial-based approach for enhancing cognitive function during ageing.


Subject(s)
Clostridiales , Cognition Disorders , Cognitive Dysfunction , Mice , Humans , Animals , Aged , Cognition Disorders/metabolism , Brain/metabolism , Aging/metabolism
3.
Int J Mol Sci ; 23(22)2022 Nov 13.
Article in English | MEDLINE | ID: mdl-36430486

ABSTRACT

Fulminant hepatitis is characterized by rapid and massive immune-mediated liver injury. Dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (DAX1; NR0B1) represses the transcription of various genes. Here, we determine whether DAX1 serves as a regulator of inflammatory liver injury induced by concanavalin A (ConA). C57BL/6J (WT), myeloid cell-specific Dax1 knockout (MKO), and hepatocyte-specific Dax1 knockout (LKO) mice received single intravenous administration of ConA. Histopathological changes in liver and plasma alanine aminotransferase and aspartate aminotransferase levels in Dax1 MKO mice were comparable with those in WT mice following ConA administration. Unlike Dax1 MKO mice, Dax1 LKO mice were greatly susceptible to ConA-induced liver injury, which was accompanied by enhanced infiltration of immune cells, particularly CD4+ and CD8+ T cells, in the liver. Factors related to T-cell recruitment, including chemokines and adhesion molecules, significantly increased following enhanced and prolonged phosphorylation of NF-κB p65 in the liver of ConA-administered Dax1 LKO mice. This is the first study to demonstrate that hepatocyte-specific DAX1 deficiency exacerbates inflammatory liver injury via NF-κB p65 activation, thereby causing T-cell infiltration by modulating inflammatory chemokines and adhesion molecules. Our results suggest DAX1 as a therapeutic target for fulminant hepatitis treatment.


Subject(s)
CD8-Positive T-Lymphocytes , Massive Hepatic Necrosis , Mice , Animals , NF-kappa B , Mice, Inbred C57BL , Hepatocytes , Signal Transduction , Concanavalin A/toxicity , CD4-Positive T-Lymphocytes
4.
Int J Mol Sci ; 23(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36233086

ABSTRACT

Acetaminophen (APAP) is a widely used analgesic and antipyretic drug, but its overdose can cause acute liver failure. The dosage-sensitive sex reversal adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (DAX-1, NR0B1), is an orphan nuclear receptor that acts as a transcriptional co-repressor of various genes. In this study, we identified the role of DAX-1 in APAP-induced liver injury using hepatocyte-specific Dax-1 knockout (Dax-1 LKO) mice. Mouse primary hepatocytes were used as a comparative in vitro study. APAP overdose led to decreased plasma alanine aminotransferase and aspartate aminotransferase levels in Dax-1 LKO mice compared to C57BL/6J (WT) controls, accompanied by reduced liver necrosis. The expression of the genes encoding the enzymes catalyzing glutathione (GSH) synthesis and metabolism and antioxidant enzymes was increased in the livers of APAP-treated Dax-1 LKO mice. The rapid recovery of GSH levels in the mitochondrial fraction of APAP-treated Dax-1 LKO mice led to reduced reactive oxygen species levels, resulting in the inhibition of the prolonged JNK activation. The hepatocyte-specific DAX-1 deficiency increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2) compared with WT controls after APAP administration. These results indicate that DAX-1 deficiency in hepatocytes protects against APAP-induced liver injury by Nrf2-regulated antioxidant defense.


Subject(s)
Antipyretics , Chemical and Drug Induced Liver Injury , DAX-1 Orphan Nuclear Receptor , NF-E2-Related Factor 2 , Acetaminophen/toxicity , Alanine Transaminase/metabolism , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Co-Repressor Proteins/metabolism , DAX-1 Orphan Nuclear Receptor/genetics , Glutathione/metabolism , Hepatocytes/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Orphan Nuclear Receptors/metabolism , Reactive Oxygen Species/metabolism
5.
Psychopharmacology (Berl) ; 238(7): 1833-1845, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33723660

ABSTRACT

Autism spectrum disorder (ASD) is a neurodevelopment disorder characterized by deficits in social interaction and restrictive, repetitive, and stereotypical patterns of behavior. However, there is no pharmacological drug that is currently used to target these core ASD symptoms. Sodium phenylbutyrate (NaPB) is a well-known long-term treatment of urea cycle disorders in children. In this study, we assessed the therapeutic effects of NaPB, which is a chemical chaperone as well as histone deacetylase inhibitor on a BTBR T + Itpr3tf/J (BTBR) mice model of ASD. We found that acute and chronic treatment of NaPB remarkably improved, not only core ASD symptoms, including repetitive behaviors and sociability deficit, but also cognitive impairment in the BTBR mice. NaPB substantially induced histone acetylation in the brain of the BTBR mice. Intriguingly, the therapeutic effects of NaPB on autistic-like behaviors, such as repetitive behaviors, impaired sociability, and cognitive deficit also showed in the valproic acid (VPA)-induced mouse model of autism. In addition, pentylenetetrazole (PTZ)-induced seizure was significantly attenuated by NaPB treatment in C57BL/6J and BTBR mice. These findings suggest that NaPB may provide a novel therapeutic approach for the treatment of patients with ASD.


Subject(s)
Autism Spectrum Disorder/drug therapy , Cognitive Dysfunction/drug therapy , Grooming/drug effects , Phenylbutyrates/therapeutic use , Social Behavior , Stereotyped Behavior/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/psychology , Brain/drug effects , Brain/physiology , Cognitive Dysfunction/psychology , Disease Models, Animal , Female , Grooming/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Phenylbutyrates/pharmacology , Stereotyped Behavior/physiology , Valproic Acid/toxicity
6.
Mol Med Rep ; 23(3)2021 03.
Article in English | MEDLINE | ID: mdl-33495840

ABSTRACT

The dopamine precursor 3,4­dihydroxyphenyl­ l­alanine (L­DOPA) is the most widely used symptomatic treatment for Parkinson's disease (PD); however, its prolonged use is associated with L­DOPA­induced dyskinesia in more than half of patients after 10 years of treatment. The present study investigated whether co­treatment with ß­Lapachone, a natural compound, and L­DOPA has protective effects in a 6­hydroxydopamine (6­OHDA)­induced mouse model of PD. Unilateral 6­OHDA­lesioned mice were treated with vehicle or ß­Lapachone (10 mg/kg/day) and L­DOPA for 11 days. Abnormal involuntary movements (AIMs) were scored on days 5 and 10. ß­Lapachone (10 mg/kg) co­treatment with L­DOPA decreased the AIMs score on both days 5 and 10. ß­Lapachone was demonstrated to have a beneficial effect on the axial and limb AIMs scores on day 10. There was no significant suppression in dopamine D1 receptor­related and ERK1/2 signaling in the DA­denervated striatum by ß­Lapachone­cotreatment with L­DOPA. Notably, ß­Lapachone­cotreatment with L­DOPA increased phosphorylation at the Ser9 site of glycogen synthase kinase 3ß (GSK­3ß), indicating suppression of GSK­3ß activity in both the unlesioned and 6­OHDA­lesioned striata. In addition, astrocyte activation was markedly suppressed by ß­Lapachone­cotreatment with L­DOPA in the striatum and substantia nigra of the unilateral 6­OHDA model. These findings suggest that ß­Lapachone cotreatment with L­DOPA therapy may have therapeutic potential for the suppression or management of the development of L­DOPA­induced dyskinesia in patients with PD.


Subject(s)
Dyskinesias , Levodopa/adverse effects , Naphthoquinones/pharmacology , Oxidopamine/adverse effects , Parkinson Disease, Secondary , Animals , Dyskinesias/drug therapy , Dyskinesias/metabolism , Dyskinesias/pathology , Levodopa/pharmacology , Male , Mice , Oxidopamine/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology
7.
J Cell Mol Med ; 24(15): 8814-8825, 2020 08.
Article in English | MEDLINE | ID: mdl-32570293

ABSTRACT

The growth arrest and DNA damage-inducible beta (Gadd45ß) protein have been associated with various cellular functions, but its role in progressive renal disease is currently unknown. Here, we examined the effect of Gadd45ß deletion on cell proliferation and apoptosis, inflammation, and renal fibrosis in an early chronic kidney disease (CKD) mouse model following unilateral ureteral obstruction (UUO). Wild-type (WT) and Gadd45ß-knockout (KO) mice underwent either a sham operation or UUO and the kidneys were sampled eight days later. A histological assay revealed that ablation of Gadd45ß ameliorated UUO-induced renal injury. Cell proliferation was higher in Gadd45ß KO mouse kidneys, but apoptosis was similar in both genotypes after UUO. Expression of pro-inflammatory cytokines after UUO was down-regulated in the kidneys from Gadd45ß KO mice, whereas UUO-mediated immune cell infiltration remained unchanged. The expression of pro-inflammatory cytokines in response to LPS stimulation decreased in bone marrow-derived macrophages from Gadd45ß KO mice compared with that in WT mice. Importantly, UUO-induced renal fibrosis was ameliorated in Gadd45ß KO mice unlike in WT mice. Gadd45ß was involved in TGF-ß signalling pathway regulation in kidney fibroblasts. Our findings demonstrate that Gadd45ß plays a crucial role in renal injury and may be a therapeutic target for the treatment of CKD.


Subject(s)
Antigens, Differentiation/genetics , Gene Deletion , Kidney Diseases/etiology , Ureteral Obstruction/complications , Animals , Antigens, Differentiation/metabolism , Apoptosis/genetics , Biomarkers , Biopsy , Cell Line , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Fibrosis , Immunohistochemistry , Inflammation , Inflammation Mediators , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta/metabolism
8.
Neuropharmacology ; 175: 108173, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32497590

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the substantia nigra, leading to motor symptoms. Despite the remarkable improvements in the management of PD in recent decades, many patients remain significantly disabled. Metformin is a primary medication for the management of type 2 diabetes. We previously showed that co-treatment with metformin and 3,4-dihydroxyphenyl-l-alanine (l-DOPA) prevented the development of l-DOPA-induced dyskinesia in a 6-hydroxydopamine (6-OHDA)-lesioned animal model of PD. However, effects of metformin on PD- and aging-induced genes in reactive astrocytes remain unknown. In this study, we assessed the effect of metformin on motor function, neuroprotection, and reactive astrocytes in the 6-OHDA-induced PD animal model. In addition, the effects of metformin on the genes expressed by specific types of astrocytes were analyzed in PD model and aged mice. Here, we showed that metformin treatment effectively improves the motor symptoms in the 6-OHDA-induced PD mouse model, whereas metformin had no effect on tyrosine hydroxylase-positive neurons. The activation of AMPK and BDNF signaling pathways was induced by metformin treatment on the 6-OHDA-lesioned side of the striatum. Metformin treatment caused astrocytes to alter reactive genes in a PD animal model. Moreover, aging-induced genes in reactive astrocytes were effectively regulated or suppressed by metformin treatment. Taken together, these results suggest that metformin should be evaluated for the treatment of Parkinson's disease and related neurologic disorders characterized by astrocyte activation.


Subject(s)
Aging/physiology , Astrocytes/drug effects , Astrocytes/physiology , Corpus Striatum/physiology , Metformin/administration & dosage , Parkinson Disease/physiopathology , Animals , Corpus Striatum/drug effects , Disease Models, Animal , Female , Male , Mice, Inbred C57BL , Motor Activity/drug effects
9.
Neuropharmacology ; 170: 108039, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32165217

ABSTRACT

The NAD(P)H:quinone oxidoreductase 1 (NQO1) gene encodes a cytosolic flavoenzyme that catalyzes the two-electron reduction of quinones to hydroquinones. A polymorphic form of NQO1 is associated with mood disorders such as schizophrenia. However, the role of NQO1 in dopaminergic system has not yet been elucidated. To determine the role of NQO1 in the dopaminergic system, we investigated pharmaco-behavioral effects of d-amphetamine using NQO1-deficienct mice. According to our comparative study involving NQO1+/+ and NQO1-/- mice, NQO1 deficiency increased d-amphetamine-induced psychomotor activity and psychological dependency compared to wild-type mice. Basal and d-amphetamine-induced dopamine levels were also enhanced by NQO1 deficiency. In NQO1-/- mice, neural activation induced by d-amphetamine was higher in dorsolateral striatum, but not in dorsomedial and ventral striata. Although protein level of CaMKIIα, which is a key player in amphetamine-induced dopamine efflux, was decreased in striata of NQO1-/- mice, phosphorylation of CaMKIIα was markedly enhanced in NQO1-/- mice compared to wild-type mice. Interestingly, experiments with pharmacological antagonist showed that D2 antagonist-induced suppression of locomotion required activation of NQO1. Moreover, the rewarding effect in response to D1 agonist was increased by NQO1 deficiency. These results suggest that striatal NQO1 is of considerable interest to understand the mechanism of dopaminergic regulation of psychiatric disorders.


Subject(s)
Corpus Striatum/metabolism , Dextroamphetamine/pharmacology , Dopamine/metabolism , NAD(P)H Dehydrogenase (Quinone)/deficiency , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Animals , Corpus Striatum/drug effects , Dopamine D2 Receptor Antagonists/pharmacology , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D2/agonists
10.
Int J Mol Med ; 45(2): 417-428, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31894253

ABSTRACT

Humulus japonicus (HJ) is a widely used herbal medicine in Asia with anti­oxidative, anti­microbial, and anti­inflammatory effects. We investigated the potential therapeutic effects of HJ in rheumatoid arthritis (RA) using a mouse model of collagen­induced arthritis (CIA) and a lipopolysaccharide­stimulated murine macrophage cell line (RAW 264.7). The CIA mice were administered 300 mg/kg HJ orally starting 3 days prior to second immunization. The clinical and histopathological findings were assessed in the paw of CIA mice. The levels of autoantibodies and inflammatory markers were determined in the plasma and cell culture supernatant, respectively. The expression at mRNA and protein levels was analyzed by reverse transcription quantitative­PCR and western blot analysis, respectively. HJ significantly decreased the gross arthritic scores and paw swelling in CIA mice. Furthermore, synovial inflammation, cartilage destruction, and bone erosion were markedly reduced by HJ. It also decreased the expression of inflammatory enzymes in both the paw of mice and RAW 264.7 cells. Moreover, the expression of genes related to all macrophages and pro­inflammatory M1 macrophage were significantly decreased, whereas the expression of anti­inflammatory M2 macrophage marker was markedly increased in the paw of HJ­treated CIA mice. In addition, HJ suppressed the levels of plasma anti­type II collagen antibody following the decreased expression of T helper type 1 (Th1) and Th2 cell­associated surface markers and cytokines in the paw. HJ also significantly inhibited the expression of IL­6 both in vitro and in vivo, followed by reduced STAT3 phosphorylation and expression in the paw of CIA mice. Finally, the expression of osteoclast­related genes was decreased in the paw of HJ­treated CIA mice. These findings suggest that HJ can play a role in suppressing the development of CIA by overall regulation of articular inflammation. This study should provide new insights into the use of HJ as a therapeutically effective natural product against RA.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Humulus , Plant Extracts/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Autoantibodies/immunology , Disease Models, Animal , Humulus/chemistry , Inflammation/drug therapy , Inflammation/immunology , Inflammation Mediators/immunology , Male , Mice , Plant Extracts/chemistry , RAW 264.7 Cells
11.
Cells ; 8(11)2019 11 14.
Article in English | MEDLINE | ID: mdl-31739636

ABSTRACT

Clusterin is a glycoprotein that is expressed in most human tissues and found in body fluids. In our previous studies we demonstrated that clusterin has a protective effect against hepatic lipid accumulation and renal fibrosis; however, the role of clusterin in hepatic fibrosis is unknown. Here, we examined whether clusterin had protective effects against hepatic fibrosis using in vitro and in vivo models. Clusterin was upregulated in the livers of human cirrhotic patients and in thioacetamide (TAA)-induced and bile duct ligation mouse models of liver fibrosis. Loss and overexpression of clusterin promoted and attenuated hepatic fibrosis after TAA injection, respectively. In addition, we found that clusterin attenuates hepatic fibrosis by inhibiting the activation of hepatic stellate cells and Smad3 signaling pathways. Thus, clusterin plays an important role in hepatic fibrosis.


Subject(s)
Clusterin/genetics , Hepatic Stellate Cells/cytology , Liver Cirrhosis/metabolism , Smad3 Protein/metabolism , Thioacetamide/adverse effects , Animals , Cell Line , Clusterin/metabolism , Disease Models, Animal , Down-Regulation , Gene Knockout Techniques , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Male , Mice , Signal Transduction , Up-Regulation
12.
Kidney Int ; 91(4): 880-895, 2017 04.
Article in English | MEDLINE | ID: mdl-28040265

ABSTRACT

Clinical prescription of cisplatin, one of the most widely used chemotherapeutic agents, is limited by its side effects, particularly tubular injury-associated nephrotoxicity. Since details of the underlying mechanisms are not fully understood, we investigated the role of pyruvate dehydrogenase kinase (PDK) in cisplatin-induced acute kidney injury. Among the PDK isoforms, PDK4 mRNA and protein levels were markedly increased in the kidneys of mice treated with cisplatin, and c-Jun N-terminal kinase activation was involved in cisplatin-induced renal PDK4 expression. Treatment with the PDK inhibitor sodium dichloroacetate (DCA) or genetic knockout of PDK4 attenuated the signs of cisplatin-induced acute kidney injury, including apoptotic morphology of the kidney tubules along with numbers of TUNEL-positive cells, cleaved caspase-3, and renal tubular injury markers. Cisplatin-induced suppression of the mitochondrial membrane potential, oxygen consumption rate, expression of electron transport chain components, cytochrome c oxidase activity, and disruption of mitochondrial morphology were noticeably improved in the kidneys of DCA-treated or PDK4 knockout mice. Additionally, levels of the oxidative stress marker 4-hydroxynonenal and mitochondrial reactive oxygen species were attenuated, whereas superoxide dismutase 2 and catalase expression and glutathione synthetase and glutathione levels were recovered in DCA-treated or PDK4 knockout mice. Interestingly, lipid accumulation was considerably attenuated in DCA-treated or PDK4 knockout mice via recovered expression of peroxisome proliferator-activated receptor-α and coactivator PGC-1α, which was accompanied by recovery of mitochondrial biogenesis. Thus, PDK4 mediates cisplatin-induced acute kidney injury, suggesting that PDK4 might be a therapeutic target for attenuating cisplatin-induced acute kidney injury.


Subject(s)
Acute Kidney Injury/prevention & control , Cisplatin , Kidney Tubules/enzymology , Protein Serine-Threonine Kinases/deficiency , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Apoptosis , Caspase 3/metabolism , Cells, Cultured , Disease Models, Animal , Energy Metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Genetic Predisposition to Disease , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Tubules/drug effects , Kidney Tubules/ultrastructure , Male , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/enzymology , Mitochondria/pathology , Organelle Biogenesis , Oxidative Stress , Phenotype , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
13.
Kidney Int ; 90(6): 1285-1297, 2016 12.
Article in English | MEDLINE | ID: mdl-27616741

ABSTRACT

The hallmark of renal tubulointerstitial fibrosis is the accumulation of myofibroblasts and extracellular matrix proteins. Fyn, a member of the Src family of kinases, has diverse biological functions including regulation of mitogenic signaling and proliferation and integrin-mediated interaction. Src family proteins promote pulmonary fibrosis by augmenting transforming growth factor-ß signaling, but their role in renal fibrosis is less understood. We observed upregulation of Fyn in a renal fibrosis model induced by unilateral ureteral obstruction. Upon ureteral obstruction, Fyn-deficient mice exhibited attenuated renal fibrosis relative to wild-type mice. Furthermore, obstruction-induced renal expression of type I collagen, fibronectin, α-smooth muscle actin, and plasminogen activator inhibitor-1 was suppressed. Pharmacologic inhibition of Fyn blocked induction of extracellular matrix proteins in kidney cell lines. Importantly, the attenuation of renal fibrosis by Fyn deficiency was not accompanied by changes in the Smad pathway. Rather, the antifibrotic effect of Fyn deficiency was associated with downregulation of signal transducer and activator of transcription 3 (STAT3). Small, interfering RNA targeting STAT3 in Fyn-deficient cells further suppressed α-smooth muscle actin expression, whereas a STAT3 activator partially restored plasminogen activator inhibitor-1 expression, indicating that STAT3 signaling is critically involved in this process. Thus, Fyn plays an important role in renal fibrosis. Hence, Fyn kinase inhibitors may be therapeutically useful against renal fibrosis.


Subject(s)
Nephrosclerosis/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , STAT3 Transcription Factor/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cadherins/metabolism , ErbB Receptors/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Nephrosclerosis/etiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/complications , src-Family Kinases/metabolism
14.
Diabetes ; 65(10): 2876-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27385159

ABSTRACT

Hepatic steatosis is associated with increased insulin resistance and tricarboxylic acid (TCA) cycle flux, but decreased ketogenesis and pyruvate dehydrogenase complex (PDC) flux. This study examined whether hepatic PDC activation by inhibition of pyruvate dehydrogenase kinase 2 (PDK2) ameliorates these metabolic abnormalities. Wild-type mice fed a high-fat diet exhibited hepatic steatosis, insulin resistance, and increased levels of pyruvate, TCA cycle intermediates, and malonyl-CoA but reduced ketogenesis and PDC activity due to PDK2 induction. Hepatic PDC activation by PDK2 inhibition attenuated hepatic steatosis, improved hepatic insulin sensitivity, reduced hepatic glucose production, increased capacity for ß-oxidation and ketogenesis, and decreased the capacity for lipogenesis. These results were attributed to altered enzymatic capacities and a reduction in TCA anaplerosis that limited the availability of oxaloacetate for the TCA cycle, which promoted ketogenesis. The current study reports that increasing hepatic PDC activity by inhibition of PDK2 ameliorates hepatic steatosis and insulin sensitivity by regulating TCA cycle anaplerosis and ketogenesis. The findings suggest PDK2 is a potential therapeutic target for nonalcoholic fatty liver disease.


Subject(s)
Fatty Liver/enzymology , Fatty Liver/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Citric Acid Cycle/genetics , Citric Acid Cycle/physiology , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Glucose/metabolism , Insulin Resistance , Lipogenesis/physiology , Liver/metabolism , Liver/pathology , Male , Malonyl Coenzyme A/metabolism , Mice , Mice, Knockout , Oxaloacetic Acid/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Dehydrogenase Complex/metabolism , Pyruvic Acid/metabolism
16.
Sci Rep ; 5: 16577, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26560812

ABSTRACT

Vascular calcification, a pathologic response to defective calcium and phosphate homeostasis, is strongly associated with cardiovascular mortality and morbidity. In this study, we have observed that pyruvate dehydrogenase kinase 4 (PDK4) is upregulated and pyruvate dehydrogenase complex phosphorylation is increased in calcifying vascular smooth muscle cells (VSMCs) and in calcified vessels of patients with atherosclerosis, suggesting that PDK4 plays an important role in vascular calcification. Both genetic and pharmacological inhibition of PDK4 ameliorated the calcification in phosphate-treated VSMCs and aortic rings and in vitamin D3-treated mice. PDK4 augmented the osteogenic differentiation of VSMCs by phosphorylating SMAD1/5/8 via direct interaction, which enhances BMP2 signaling. Furthermore, increased expression of PDK4 in phosphate-treated VSMCs induced mitochondrial dysfunction followed by apoptosis. Taken together, our results show that upregulation of PDK4 promotes vascular calcification by increasing osteogenic markers with no adverse effect on bone formation, demonstrating that PDK4 is a therapeutic target for vascular calcification.


Subject(s)
Protein Kinases/genetics , Protein Kinases/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Vascular Calcification/metabolism , Animals , Apoptosis/genetics , Biomarkers , Bone Remodeling/genetics , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Osteogenesis/genetics , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry
17.
Arterioscler Thromb Vasc Biol ; 35(11): 2384-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26404484

ABSTRACT

OBJECTIVE: Vascular calcification which refers to ectopic mineralization in vascular cells is associated with several conditions, such as chronic kidney disease, atherosclerosis, and diabetes mellitus. Estrogen-related receptor (ERR)γ is a member of the orphan nuclear receptor superfamily, which plays diverse roles in regulating homeostatic and metabolic processes. However, the role of ERRγ in vascular calcification has not been investigated to date. The aim of the present study was to examine the role of ERRγ in vascular calcification. APPROACH AND RESULTS: Vascular calcification was induced by treating rat aortic vascular smooth muscle cells with calcification medium. ERRγ expression in vascular smooth muscle cells was induced during calcification medium-induced vascular calcification. Adenovirus-mediated overexpression of ERRγ in vascular smooth muscle cells resulted in the upregulation of the expression of osteogenic genes, including runt-related transcription factor 2, osteopontin, and Msx2, and the downregulation of α-smooth muscle actin. Adenovirus-mediated overexpression of ERRγ induced bone morphogenetic protein 2 (BMP2) expression, leading to increased phosphorylation of the intracellular BMP2 effector proteins SMAD1/5/8. Collectively, these data suggested that ERRγ promotes dedifferentiation of vascular smooth muscle cells to an osteogenic phenotype during the vascular calcification process. Inhibition of endogenous ERRγ expression or activity using a specific siRNA or the selective inverse agonist GSK5182 attenuated vascular calcification and osteogenic gene expression in vitro and in vivo. CONCLUSIONS: The present results indicate that ERRγ plays a key role in vascular calcification by upregulating the BMP2 signaling pathway, suggesting that inhibition of ERRγ is a potential therapeutic strategy for the prevention of vascular calcification.


Subject(s)
Aortic Diseases/metabolism , Bone Morphogenetic Protein 2/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis , Receptors, Estrogen/metabolism , Vascular Calcification/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Binding Sites , Bone Morphogenetic Protein 2/genetics , Cell Dedifferentiation , Cell Line , Disease Models, Animal , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Promoter Regions, Genetic , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Signal Transduction , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Time Factors , Transfection , Up-Regulation , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/prevention & control
18.
Biochem Biophys Res Commun ; 460(2): 457-63, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25796334

ABSTRACT

Ischemic retinopathies causing overexpression of pro-angiogenic factors, including vascular endothelial growth factor (VEGF), are the most common cause of blindness. Thus, understanding the pathophysiology of targetable pathways that regulate retinal VEGF is of great interest. A conserved binding site for estrogen-related receptor γ (ERRγ) has been identified in the promoter of the Vegfa gene. ERRγ is a constitutively active orphan nuclear receptor and its expression is increased by hypoxic stimuli in metabolically active tissues. This study evaluated the role of ERRγ in the ischemic retina and the anti-VEGF potential of GSK5182, a selective inverse agonist of ERRγ. In an oxygen-induced retinopathy (OIR) mouse model, immunohistochemistry showed significantly increased ERRγ expression in the ganglion cell layer at postnatal day (P) 17. In a ganglion cell line (RGC-5), mRNA and protein levels of ERRγ were increased by desferrioxamine treatment and hypoxic conditions (1% O2). Transient transfection of RGC-5 cells revealed that ERRγ regulated Vegfa expression and this was inhibited by GSK5182. Intravitreal injection of GSK5182 into the OIR model at P14 inhibited retinal Vegfa mRNA expression at P17. GSK5182 suppresses hypoxia-induced VEGF expression via ERRγ; therefore, ERRγ could be a treatment target for ischemic retinopathies.


Subject(s)
Hypoxia/metabolism , Receptors, Estrogen/biosynthesis , Retina/pathology , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Cell Line , Mice , Mice, Inbred C57BL , Rats , Reverse Transcriptase Polymerase Chain Reaction
19.
Vascul Pharmacol ; 63(1): 29-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25135648

ABSTRACT

Dimethyl fumarate (DMF) has several pharmacological benefits including immunomodulation and prevention of fibrosis, which are dependent on the NF-E2-related factor 2 (Nrf2) antioxidant pathways. Therefore, we hypothesized that DMF could attenuate vascular calcification via Nrf2 activation. Vascular calcification induced by hyperphosphataemia was significantly inhibited by DMF in vascular smooth muscle cells (VSMCs) in a dose-dependent manner. DMF-mediated Nrf2 upregulation was accompanied by the reduced expressions of genes related with osteoblast-like phenotype based on promoter activity, mRNA and protein expression, and von Kossa staining. Likewise, Nrf2 overexpression significantly decreased the formation of calcium deposit similar to the level of osteogenic staining in VSMCs, and DMF with Nrf2 knockdown failed to attenuate hyperphosphatemia induced vascular calcification. Furthermore, DMF significantly attenuated the calcification of ex vivo ring culture from both rat common carotid artery and mouse thoracic aorta as well as in vivo mouse model of Vitamin D3-induced calcification consistent with the increased Nrf2 protein levels in early stage of calcification by DMF. In conclusion, our data support that DMF stimulates Nrf2 activity to attenuate hyperphosphatamia in vitro or Vitamin D3-induced in vivo vascular calcification, which would be a beneficial effect on vascular diseases induced by oxidative stress such as vascular calcification.


Subject(s)
Fumarates/pharmacology , Muscle, Smooth, Vascular/drug effects , NF-E2-Related Factor 2/metabolism , Vascular Calcification/drug therapy , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Calcium/metabolism , Carotid Artery, Common/drug effects , Carotid Artery, Common/pathology , Dimethyl Fumarate , Dose-Response Relationship, Drug , Fumarates/administration & dosage , Gene Expression Regulation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Oxidative Stress/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects , Vascular Calcification/pathology
20.
Int J Cardiol ; 175(3): 515-21, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25017905

ABSTRACT

BACKGROUND: Retinoic acid-related orphan receptor α (RORα) has been implicated in the progression of atherosclerosis, but its role in the proliferation of vascular smooth muscle cells (vSMCs) has not been fully examined. We previously reported that RORα activates AMP-activated protein kinase (AMPK), which is associated with the suppression of vSMC proliferation. Therefore, we investigated the suppressive function of RORα on the proliferation of vSMCs and the molecular mechanisms involved. RESULTS: First, RORα and its activator, cholesterol sulfate (CS), induced the activation of AMPK in both human aortic SMCs and rat A7r5 cells, which was accompanied by the suppression of mammalian target of rapamycin (mTOR) and p70 ribosomal protein S6 kinase 1. Second, RORα and CS modulated the expression of cell-cycle-regulating factors, such as p53, p27, and cyclin D in vSMCs. Consistent with this, the overexpression of RORα or CS treatment suppressed the proliferation of human aortic SMCs and rat A7r5 cells, possibly through G1 arrest. RORα and CS also inhibited the migration of A7r5 cells in two-dimensional and three-dimensional cell migration assays. Finally, we demonstrated that the infusion of adenovirus encoding RORα into arteries suppressed neointima formation after balloon injury in rats. CONCLUSION: These results demonstrate that RORα inhibits vSMC proliferation through AMPK-induced mTOR suppression, and suggest that RORα is a therapeutic target for the cardiovascular diseases associated with vSMC proliferation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Proliferation/physiology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Nuclear Receptor Subfamily 1, Group F, Member 1/administration & dosage , Animals , Cell Line , Cell Proliferation/drug effects , Enzyme Activation/physiology , Humans , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...