Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cardiovasc Drugs Ther ; 36(4): 619-632, 2022 08.
Article in English | MEDLINE | ID: mdl-33886003

ABSTRACT

PURPOSE: Although the cardioprotective benefits of sodium-glucose cotransporter 2 (SGLT2) inhibitors are now widely appreciated, the mechanisms underlying these benefits remain unresolved. Tumor necrosis factor receptor superfamily member 12a (Tnfrsf12a) is a receptor for tumor necrosis factor superfamily member 12 (Tnfsf12). Tnfrsf12a is highly inducible and plays a key role in the development of cardiac hypertrophy and heart failure. Here we set out to determine if SGLT2 inhibition affects the Tnfsf12/Tnfrsf12a system in the stressed myocardium. METHODS: C57BL/6N mice that had undergone sham or transverse aortic constriction (TAC) surgery were treated with either the SGLT2 inhibitor empagliflozin (400 mg/kg diet; 60-65 mg/kg/day) or standard chow alone and were followed for 8 weeks. Tnfrsf12a expression in mouse hearts was assessed by in situ hybridization, qRT-PCR, and immunoblotting. RESULTS: Left ventricular (LV) mass, end-systolic volume, and end-diastolic volume were all increased in TAC mice and were significantly lower with empagliflozin. Myocyte hypertrophy and interstitial fibrosis in TAC hearts were similarly attenuated with empagliflozin. Tnfrsf12a expression was upregulated in mouse hearts following TAC surgery but not in the hearts of empagliflozin-treated mice. In cultured cardiomyocytes, Tnfrsf12a antagonism attenuated the increase in cardiomyocyte size that was induced by phenylephrine. CONCLUSION: Empagliflozin attenuates LV enlargement in mice with hypertrophic heart failure. This effect may be mediated, at least in part, by a reduction in loading conditions which limits upregulation of the inducible, proinflammatory, and prohypertrophic TNF superfamily receptor, Tnfrsf12a. Disruption of the Tnfsf12/Tnfrsf12a feed forward system may contribute to the cardioprotective benefits of SGLT2 inhibition.


Subject(s)
Heart Failure , Hypertrophy, Left Ventricular , TWEAK Receptor/metabolism , Animals , Benzhydryl Compounds , Glucosides , Heart Failure/metabolism , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/prevention & control , Mice , Mice, Inbred C57BL , Myocytes, Cardiac , Sodium-Glucose Transporter 2/metabolism , Ventricular Remodeling
2.
Circulation ; 142(23): 2240-2258, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33070627

ABSTRACT

BACKGROUND: Cardiac hypertrophy is a key biological response to injurious stresses such as pressure overload and, when excessive, can lead to heart failure. Innate immune activation by danger signals, through intracellular pattern recognition receptors such as nucleotide-binding oligomerization domain 1 (Nod1) and its adaptor receptor-interacting protein 2 (RIP2), might play a major role in cardiac remodeling and progression to heart failure. We hypothesize that Nod1/RIP2 are major contributors to cardiac hypertrophy, but may not be sufficient to fully express the phenotype alone. METHODS: To elucidate the contribution of Nod1/RIP2 signaling to cardiac hypertrophy, we randomized Nod1-/-, RIP2-/-, or wild-type mice to transverse aortic constriction or sham operations. Cardiac hypertrophy, fibrosis, and cardiac function were examined in these mice. RESULTS: Nod1 and RIP2 proteins were upregulated in the heart after transverse aortic constriction, and this was paralleled by increased expression of mitochondrial proteins, including mitochondrial antiviral signaling protein (MAVS). Nod1-/- and RIP2-/- mice subjected to transverse aortic constriction exhibited better survival, improved cardiac function, and decreased cardiac hypertrophy. Downstream signal transduction pathways that regulate inflammation and fibrosis, including NF (nuclear factor) κB and MAPK (mitogen-activated protein kinase)-GATA4/p300, were reduced in both Nod1-/- and RIP2-/- mice after transverse aortic constriction compared with wild-type mice. Coimmunoprecipitation of extracted cardiac proteins and confocal immunofluorescence microscopy showed that Nod1/RIP2 interaction was robust and that this complex also included MAVS as an essential component. Suppression of MAVS expression attenuated the complex formation, NF κB signaling, and myocyte hypertrophy. Interrogation of mitochondrial function compared in the presence or ablation of MAVS revealed that MAVS serves to suppress mitochondrial energy output and mediate fission/fusion related dynamic changes. The latter is possibly linked to mitophagy during cardiomyocytes stress, which may provide an intriguing link between innate immune activation and mitochondrial energy balance under stress or injury conditions. CONCLUSIONS: We have identified that innate immune Nod1/RIP2 signaling is a major contributor to cardiac remodeling after stress. This process is critically joined by and regulated through the mitochondrial danger signal adapter MAVS. This novel complex coordinates remodeling, inflammatory response, and mitochondrial energy metabolism in stressed cardiomyocytes. Thus, Nod1/RIP2/MAVS signaling complex may represent an attractive new therapeutic approach toward heart failure.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cardiomegaly/immunology , Energy Metabolism/physiology , Immunity, Innate/physiology , Nod1 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Cardiomegaly/metabolism , Cardiomegaly/pathology , Female , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction/physiology
3.
Int J Mol Sci ; 21(16)2020 Aug 11.
Article in English | MEDLINE | ID: mdl-32796688

ABSTRACT

Despite a similar mechanism of action underlying their glucose-lowering effects in type 2 diabetes, dipeptidyl peptidase-4 (DPP-4) inhibitors have diverse molecular structures, raising the prospect of agent-specific, glucose-independent actions. To explore the issue of possible DPP-4 inhibitor cardiac heterogeneity, we perfused different DPP-4 inhibitors to beating mouse hearts ex vivo, at concentrations equivalent to peak plasma levels achieved in humans with standard dosing. We studied male and female mice, young non-diabetic mice, and aged diabetic high fat diet-fed mice and observed that linagliptin enhanced recovery after ischemia-reperfusion, whereas sitagliptin, alogliptin, and saxagliptin did not. DPP-4 transcripts were not detected in adult mouse cardiomyocytes by RNA sequencing and the addition of linagliptin caused ≤0.2% of cardiomyocyte genes to be differentially expressed. In contrast, incubation of C166 endothelial cells with linagliptin induced cell signaling characterized by phosphorylation of Akt and endothelial nitric oxide synthase, whereas the nitric oxide (NO) donor, S-nitroso-N-acetylpenicillamine increased serine 16 phosphorylation of the calcium regulatory protein, phospholamban in cardiomyocytes. Furthermore, linagliptin increased cardiomyocyte cGMP when cells were co-cultured with C166 endothelial cells, but not when cardiomyocytes were cultured alone. Thus, at a concentration comparable to that achieved in patients, linagliptin has direct effects on mouse hearts. The effects of linagliptin on cardiomyocytes are likely to be either off-target or indirect, mediated through NO generation by the adjacent cardiac endothelium.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Heart/physiology , Linagliptin/pharmacology , Myocardial Contraction/drug effects , Aging/pathology , Animals , Calcium Signaling/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/physiopathology , Diet, High-Fat , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Female , Heart/drug effects , Humans , Linagliptin/therapeutic use , Male , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Perfusion , Phosphorylation/drug effects , Phosphoserine/metabolism
4.
Diabetologia ; 62(11): 2129-2142, 2019 11.
Article in English | MEDLINE | ID: mdl-31399844

ABSTRACT

AIMS/HYPOTHESIS: Long non-coding RNAs (lncRNAs) are garnering increasing attention for their putative roles in the pathogenesis of chronic diseases, including diabetic kidney disease (DKD). However, much about in vivo lncRNA functionality in the adult organism remains unclear. To better understand lncRNA regulation and function in DKD, we explored the effects of the modular scaffold lncRNA HOTAIR (HOX antisense intergenic RNA), which approximates chromatin modifying complexes to their target sites on the genome. METHODS: Experiments were performed in human kidney tissue, in mice with streptozotocin-induced diabetes, the db/db mouse model of type 2 diabetes, podocyte-specific Hotair knockout mice and conditionally immortalised mouse podocytes. RESULTS: HOTAIR was observed to be expressed by several kidney cell-types, including glomerular podocytes, in both human and mouse kidneys. However, knockout of Hotair from podocytes had almost no effect on kidney structure, function or ultrastructure. Glomerular HOTAIR expression was found to be increased in human DKD, in the kidneys of mice with streptozotocin-induced diabetes and in the kidneys of db/db mice. Likewise, exposure of cultured mouse podocytes to high glucose caused upregulation of Hotair expression, which occurred in a p65-dependent manner. Although HOTAIR expression was upregulated in DKD and in high glucose-exposed podocytes, its knockout did not alter the development of kidney damage in diabetic mice. Rather, in a bioinformatic analysis of human kidney tissue, HOTAIR expression closely paralleled the expression of its genic neighbour, HOXC11, which is important to developmental patterning but which has an uncertain role in the adult kidney. CONCLUSIONS/INTERPRETATION: Many lncRNAs have been found to bind to the same chromatin modifying complexes. Thus, there is likely to exist sufficient redundancy in the system that the biological effects of dysregulated lncRNAs in kidney disease may often be inconsequential. The example of the archetypal scaffold lncRNA, HOTAIR, illustrates how lncRNA dysregulation may be a bystander in DKD without necessarily contributing to the pathogenesis of the condition. In the absence of in vivo validation, caution should be taken before ascribing major functional roles to single lncRNAs in the pathogenesis of chronic diseases.


Subject(s)
Diabetic Nephropathies/metabolism , Gene Expression Regulation , RNA, Long Noncoding/metabolism , Animals , Body Patterning , Chromatin/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Homeodomain Proteins/metabolism , Humans , In Situ Hybridization , Kidney Glomerulus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes/cytology , Podocytes/metabolism , RNA, Long Noncoding/genetics
5.
J Am Soc Nephrol ; 28(9): 2641-2653, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28424277

ABSTRACT

The nonreceptor kinase Janus kinase 2 (JAK2) has garnered attention as a promising therapeutic target for the treatment of CKD. However, being ubiquitously expressed in the adult, JAK2 is also likely to be necessary for normal organ function. Here, we investigated the phenotypic effects of JAK2 deficiency. Mice in which JAK2 had been deleted from podocytes exhibited an elevation in urine albumin excretion that was accompanied by increased podocyte autophagosome fractional volume and p62 aggregation, which are indicative of impaired autophagy completion. In cultured podocytes, knockdown of JAK2 similarly impaired autophagy and led to downregulation in the expression of lysosomal genes and decreased activity of the lysosomal enzyme, cathepsin D. Because transcription factor EB (TFEB) has recently emerged as a master regulator of autophagosome-lysosome function, controlling the expression of several of the genes downregulated by JAK2 knockdown, we questioned whether TFEB is regulated by JAK2. In immortalized mouse podocytes, JAK2 knockdown decreased TFEB promoter activity, expression, and nuclear localization. In silico analysis and chromatin immunoprecipitation assays revealed that the downstream mediator of JAK2 signaling STAT1 binds to the TFEB promoter. Finally, overexpression of TFEB in JAK2-deficient podocytes reversed lysosomal dysfunction and restored albumin permselectivity. Collectively, these observations highlight the homeostatic actions of JAK2 in podocytes and the importance of TFEB to autophagosome-lysosome function in these cells. These results also raise the possibility that therapeutically modulating TFEB activity may improve podocyte health in glomerular disease.


Subject(s)
Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Janus Kinase 2/genetics , Podocytes/metabolism , Albuminuria/genetics , Animals , Autophagosomes/ultrastructure , Cathepsin D/metabolism , Cells, Cultured , Computer Simulation , Down-Regulation , Gene Knockdown Techniques , Janus Kinase 2/deficiency , Janus Kinase 2/metabolism , Kidney Glomerulus/cytology , Lysosomes/ultrastructure , Male , Mice , Microtubule-Associated Proteins/metabolism , Peptides/metabolism , Phenotype , Podocytes/ultrastructure , RNA, Messenger/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
6.
Circulation ; 128(14): 1542-54, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24030499

ABSTRACT

BACKGROUND: Viral myocarditis follows a fatal course in ≈30% of patients. Interleukin-1 receptor-associated kinase 4 (IRAK4), a major nodal signal transducer in innate immunity, can play a pivotal role in host inflammatory response. We sought to determine how IRAK4 modulates inflammation and outcome in a mouse model of viral myocarditis. METHODS AND RESULTS: Myocarditis was induced after intraperitoneal inoculation of coxsackievirus B3 into C57Bl/6 IRAK4-deficient mice and their littermate controls. Mortality and viral proliferation were markedly reduced in IRAK4(-/-) mice compared with their IRAK4(+/+) littermates. Disease resistance of IRAK4(-/-) mice paralleled increased amounts of protective heart-infiltrating CCR5(+) monocytes/macrophages and enhanced interferon-α and interferon-γ production 2 days after infection. Competitive bone marrow chimera demonstrated that intact IRAK4 function inhibited heart-specific migration of bone marrow-derived CCR5(+) cells. Mechanistically, lack of IRAK4 resulted in interferon regulatory factor 5 homodimerization via reduced melanoma differentiation-associated protein 5 degradation and enhanced Stat1 and Stat5 phosphorylation. Consequently, antiviral interferon-α and interferon-γ production, as well as CCR5(+) cell recruitment, increased, whereas the overall proinflammatory response was drastically reduced in the absence of IRAK4. CONCLUSIONS: Innate immunity signal transducer IRAK4 exacerbates viral myocarditis through inhibition of interferon production and reduced mobilization of protective CCR5(+) monocytes/macrophages to the heart. The combination of IRAK4 inhibitors and antiviral adjuvants may become an attractive therapeutic approach against viral myocarditis in the future.


Subject(s)
CD11b Antigen/analysis , Coxsackievirus Infections/immunology , Interferons/biosynthesis , Interleukin-1 Receptor-Associated Kinases/physiology , Monocytes/physiology , Myocarditis/immunology , Receptors, CCR5/analysis , Adoptive Transfer , Animals , Cell Movement/physiology , Chemokine CCL5/deficiency , Chemokine CCL5/physiology , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/virology , DEAD-box RNA Helicases/metabolism , Dimerization , Disease Resistance , Enterovirus B, Human/physiology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/physiopathology , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/metabolism , Interferon-Induced Helicase, IFIH1 , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/physiopathology , Myocarditis/virology , Primary Immunodeficiency Diseases , Protein Processing, Post-Translational , Radiation Chimera , Receptors, CCR5/deficiency , Receptors, CCR5/physiology , STAT1 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Virus Replication
7.
Basic Res Cardiol ; 109(3): 411, 2014 May.
Article in English | MEDLINE | ID: mdl-24763933

ABSTRACT

c-Cbl-associated protein (CAP), also called Sorbs1 or ponsin, has been described as an essential adapter protein in the insulin-signalling pathway. Here, we describe for the first time a unique protective role for CAP in viral myocarditis. Mortality and heart failure development were increased in CAP(-/-) mice compared to CAP(+/+) littermates after Coxsackievirus (CVB3) infection. Mechanistically, CAP protected from tissue apoptosis because of reduced CD8(+) T and natural killer cell cytotoxicity. Despite reduced cytotoxic elimination of CVB3-infected cells in CAP(+/+) hearts, however, CAP enhanced interferon regulatory factor 3 (IRF3)-dependent antiviral type I interferon production and decreased viral proliferation in vitro by binding to the cytoplasmic RIG-I-like receptor melanoma differentiation-associated protein 5 (MDA5). Taken together, these findings reveal a novel modulatory role for CAP in the heart as a key protein stabilizing antiviral type I interferon production, while protecting from excessive cytotoxic responses. Our study will help to define future strategies to develop treatments to limit detrimental responses during viral heart inflammation.


Subject(s)
Apoptosis , Coxsackievirus Infections/prevention & control , Enterovirus B, Human/immunology , Interferon Type I/metabolism , Microfilament Proteins/metabolism , Myocarditis/prevention & control , Myocardium/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , DEAD-box RNA Helicases/metabolism , Disease Models, Animal , Enterovirus B, Human/genetics , Enterovirus B, Human/growth & development , Enterovirus B, Human/pathogenicity , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1 , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Myeloid Differentiation Factor 88/metabolism , Myocarditis/genetics , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/virology , Myocardium/immunology , Myocardium/pathology , Time Factors , Virus Replication
8.
Dis Model Mech ; 16(9)2023 09 01.
Article in English | MEDLINE | ID: mdl-37655466

ABSTRACT

Epigenetic processes have emerged as important modulators of kidney health and disease. Here, we studied the role of KDM6A (a histone demethylase that escapes X-chromosome inactivation) in kidney tubule epithelial cells. We initially observed an increase in tubule cell Kdm6a mRNA in male mice with unilateral ureteral obstruction (UUO). However, tubule cell knockout of KDM6A had relatively minor consequences, characterized by a small reduction in apoptosis, increase in inflammation and downregulation of the peroxisome proliferator-activated receptor (PPAR) signaling pathway. In proximal tubule lineage HK-2 cells, KDM6A knockdown decreased PPARγ coactivator-1α (PGC-1α) protein levels and mRNA levels of the encoding gene, PPARGC1A. Tubule cell Kdm6a mRNA levels were approximately 2-fold higher in female mice than in male mice, both under sham and UUO conditions. However, kidney fibrosis after UUO was similar in both sexes. The findings demonstrate Kdm6a to be a dynamically regulated gene in the kidney tubule, varying in expression levels by sex and in response to injury. Despite the context-dependent variation in Kdm6a expression, knockout of tubule cell KDM6A has subtle (albeit non-negligible) effects in the adult kidney, at least in males.


Subject(s)
Histone Demethylases , Kidney , Ureteral Diseases , Animals , Female , Male , Mice , Apoptosis , Kidney Tubules , RNA, Messenger/genetics , Ureteral Diseases/genetics , Ureteral Diseases/metabolism
9.
Br J Pharmacol ; 180(22): 2898-2915, 2023 11.
Article in English | MEDLINE | ID: mdl-37115600

ABSTRACT

BACKGROUND AND PURPOSE: Activated fibroblasts deposit fibrotic matrix in chronic kidney disease (CKD) and G-protein coupled receptors (GPCRs) are the most druggable therapeutic targets. Here, we set out to establish a transcriptional profile that identifies activated kidney fibroblasts and the GPCRs that they express. EXPERIMENTAL APPROACH: RNA sequencing and single cell qRT-PCR were performed on mouse kidneys after unilateral ureteral obstruction (UUO). Candidate expression was evaluated in mice with UUO or diabetes or injected with adriamycin or folic acid. Intervention studies were conducted in mice with diabetes or UUO. Correlative histology was performed in human kidney tissue. KEY RESULTS: Transcription factor 21 (Tcf21)+ cells that expressed 2 or 3 of Postn, Acta2 and Pdgfra were highly enriched for fibrogenic genes and were defined as activated kidney fibroblasts. Tcf21+ α-smooth muscle actin (α-SMA)+ interstitial cells accumulated in kidneys of mice with UUO or diabetes or injected with adriamycin or folic acid, whereas renin-angiotensin system blockade attenuated increases in Tcf21 in diabetic mice. Fifty-six GPCRs were up-regulated in single Tcf21+ kidney fibroblasts, the most up-regulated being Adgra2 and S1pr3. Adenosine receptors, Adora2a/2b, were up-regulated in Tcf21+ fibroblasts and the adenosine receptor antagonist, caffeine decreased Tcf21 upregulation and kidney fibrosis in UUO mice. TCF21, ADGRA2, S1PR3 and ADORA2A/2B were each detectable in α-SMA+ interstitial cells in human kidney samples. CONCLUSION AND IMPLICATIONS: Tcf21 is a marker of kidney fibroblasts that are enriched for fibrogenic genes in CKD. Further analysis of the GPCRs expressed by these cells may identify new targets for treating CKD. LINKED ARTICLES: This article is part of a themed issue on Translational Advances in Fibrosis as a Therapeutic Target. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.22/issuetoc.


Subject(s)
Diabetes Mellitus, Experimental , Kidney Diseases , Renal Insufficiency, Chronic , Ureteral Obstruction , Animals , Humans , Mice , Basic Helix-Loop-Helix Transcription Factors/metabolism , Diabetes Mellitus, Experimental/metabolism , Doxorubicin/pharmacology , Fibroblasts/metabolism , Fibrosis , Folic Acid/metabolism , Folic Acid/pharmacology , Folic Acid/therapeutic use , Kidney , Kidney Diseases/metabolism , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Renal Insufficiency, Chronic/metabolism , Transcription Factors/metabolism , Ureteral Obstruction/metabolism
10.
J Clin Invest ; 133(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-37115698

ABSTRACT

Inflammation promotes adverse ventricular remodeling, a common antecedent of heart failure. Here, we set out to determine how inflammatory cells affect cardiomyocytes in the remodeling heart. Pathogenic cardiac macrophages induced an IFN response in cardiomyocytes, characterized by upregulation of the ubiquitin-like protein IFN-stimulated gene 15 (ISG15), which posttranslationally modifies its targets through a process termed ISGylation. Cardiac ISG15 is controlled by type I IFN signaling, and ISG15 or ISGylation is upregulated in mice with transverse aortic constriction or infused with angiotensin II; rats with uninephrectomy and DOCA-salt, or pulmonary artery banding; cardiomyocytes exposed to IFNs or CD4+ T cell-conditioned medium; and ventricular tissue of humans with nonischemic cardiomyopathy. By nanoscale liquid chromatography-tandem mass spectrometry, we identified the myofibrillar protein filamin-C as an ISGylation target. ISG15 deficiency preserved cardiac function in mice with transverse aortic constriction and led to improved recovery of mouse hearts ex vivo. Metabolomics revealed that ISG15 regulates cardiac amino acid metabolism, whereas ISG15 deficiency prevented misfolded filamin-C accumulation and induced cardiomyocyte autophagy. In sum, ISG15 upregulation is a feature of pathological ventricular remodeling, and protein ISGylation is an inflammation-induced posttranslational modification that may contribute to heart failure development by altering cardiomyocyte protein turnover.


Subject(s)
Cytokines , Heart Failure , Humans , Rats , Mice , Animals , Cytokines/genetics , Cytokines/metabolism , Filamins , Ventricular Remodeling/genetics , Heart Failure/metabolism , Inflammation , Ubiquitins/genetics
11.
Circulation ; 121(24): 2624-34, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20530002

ABSTRACT

BACKGROUND: Coxsackievirus B3 infection is an excellent model of human myocarditis and dilated cardiomyopathy. Cardiac injury is caused either by a direct cytopathic effect of the virus or through immune-mediated mechanisms. Regulatory T cells (Tregs) play an important role in the negative modulation of host immune responses and set the threshold of autoimmune activation. This study was designed to test the protective effects of Tregs and to determine the underlying mechanisms. METHODS AND RESULTS: Carboxyfluorescein diacetate succinimidyl ester-labeled Tregs or naïve CD4(+) T cells were injected intravenously once every 2 weeks 3 times into mice. The mice were then challenged with intraperitoneal coxsackievirus B3 immediately after the last cell transfer. Transfer of Tregs showed higher survival rates than transfer of CD4(+) T cells (P=0.0136) but not compared with the PBS injection group (P=0.0589). Interestingly, Tregs also significantly decreased virus titers and inflammatory scores in the heart. Transforming growth factor-beta and phosphorylated AKT were upregulated in Tregs-transferred mice and coxsackie-adenovirus receptor expression was decreased in the heart compared with control groups. Transforming growth factor-beta decreased coxsackie-adenovirus receptor expression and inhibited coxsackievirus B3 infection in HL-1 cells and neonatal cardiac myocytes. Splenocytes collected from Treg-, CD4(+) T-cell-, and PBS-treated mice proliferated equally when stimulated with heat-inactivated virus, whereas in the Treg group, the proliferation rate was reduced significantly when stimulated with noninfected heart tissue homogenate. CONCLUSIONS: Adoptive transfer of Tregs protected mice from coxsackievirus B3-induced myocarditis through the transforming growth factor beta-coxsackie-adenovirus receptor pathway and thus suppresses the immune response to cardiac tissue, maintaining the antiviral immune response.


Subject(s)
Enterovirus B, Human/physiology , Myocarditis/physiopathology , Myocarditis/virology , Receptors, Cytoplasmic and Nuclear/metabolism , T-Lymphocytes, Regulatory/physiology , Transforming Growth Factor beta/metabolism , Adaptive Immunity/physiology , Animals , Cells, Cultured , Constitutive Androstane Receptor , Disease Models, Animal , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Myocarditis/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , T-Lymphocytes, Regulatory/transplantation , Tumor Necrosis Factor-alpha/metabolism
12.
J Clin Invest ; 118(3): 879-93, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18292803

ABSTRACT

Chromatin remodeling, particularly histone acetylation, plays a critical role in the progression of pathological cardiac hypertrophy and heart failure. We hypothesized that curcumin, a natural polyphenolic compound abundant in the spice turmeric and a known suppressor of histone acetylation, would suppress cardiac hypertrophy through the disruption of p300 histone acetyltransferase-dependent (p300-HAT-dependent) transcriptional activation. We tested this hypothesis using primary cultured rat cardiac myocytes and fibroblasts as well as two well-established mouse models of cardiac hypertrophy. Curcumin blocked phenylephrin-induced (PE-induced) cardiac hypertrophy in vitro in a dose-dependent manner. Furthermore, curcumin both prevented and reversed mouse cardiac hypertrophy induced by aortic banding (AB) and PE infusion, as assessed by heart weight/BW and lung weight/BW ratios, echocardiographic parameters, and gene expression of hypertrophic markers. Further investigation demonstrated that curcumin abrogated histone acetylation, GATA4 acetylation, and DNA-binding activity through blocking p300-HAT activity. Curcumin also blocked AB-induced inflammation and fibrosis through disrupting p300-HAT-dependent signaling pathways. Our results indicate that curcumin has the potential to protect against cardiac hypertrophy, inflammation, and fibrosis through suppression of p300-HAT activity and downstream GATA4, NF-kappaB, and TGF-beta-Smad signaling pathways.


Subject(s)
Cardiomegaly/prevention & control , Curcumin/pharmacology , Enzyme Inhibitors/pharmacology , Acetylation , Animals , Curcumin/therapeutic use , DNA/metabolism , Fibrosis , GATA4 Transcription Factor/metabolism , Histone Deacetylase Inhibitors , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , p300-CBP Transcription Factors/antagonists & inhibitors
13.
Circ Res ; 104(7): 896-904, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19246681

ABSTRACT

Gelsolin, a calcium-regulated actin severing and capping protein, is highly expressed in murine and human hearts after myocardial infarction and is associated with progression of heart failure in humans. The biological role of gelsolin in cardiac remodeling and heart failure progression after injury is not defined. To elucidate the contribution of gelsolin in these processes, we randomly allocated gelsolin knockout mice (GSN(-/-)) and wild-type littermates (GSN(+/+)) to left anterior descending coronary artery ligation or sham surgery. We found that GSN(-/-) mice have a surprisingly lower mortality, markedly reduced hypertrophy, smaller late infarct size, less interstitial fibrosis, and improved cardiac function when compared with GSN(+/+) mice. Gene expression and protein analysis identified significantly lower levels of deoxyribonuclease (DNase) I and reduced nuclear translocation and biological activity of DNase I in GSN(-/-) mice. Absence of gelsolin markedly reduced DNase I-induced apoptosis. The association of hypoxia-inducible factor (HIF)-1alpha with gelsolin and actin filaments cleaved by gelsolin may contribute to the higher activation of DNase. The expression pattern of HIF-1alpha was similar to that of gelsolin, and HIF-1alpha was detected in the gelsolin complex by coprecipitation and HIF-1alpha bound to the promoter of DNase I in both gel-shift and promoter activity assays. Furthermore, the phosphorylation of Akt at Ser473 and expression of Bcl-2 were significantly increased in GSN(-/-) mice, suggesting that gelsolin downregulates prosurvival factors. Our investigation concludes that gelsolin is an important contributor to heart failure progression through novel mechanisms of HIF-1alpha and DNase I activation and downregulation of antiapoptotic survival factors. Gelsolin inhibition may form a novel target for heart failure therapy.


Subject(s)
Apoptosis , Deoxyribonuclease I/metabolism , Gelsolin/metabolism , Heart Failure/enzymology , Myocardial Infarction/enzymology , Myocardium/enzymology , Ventricular Remodeling , Actin Cytoskeleton/metabolism , Animals , Caspases/metabolism , Deoxyribonuclease I/genetics , Disease Models, Animal , Disease Progression , Enzyme Activation , Fibrosis , Gelsolin/deficiency , Gelsolin/genetics , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Promoter Regions, Genetic , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Time Factors , Up-Regulation , Ventricular Function, Left
14.
Diabetes ; 70(3): 759-771, 2021 03.
Article in English | MEDLINE | ID: mdl-33310740

ABSTRACT

The causes of the increased risk of severe coronavirus disease 2019 (COVID-19) in people with diabetes are unclear. It has been speculated that renin-angiotensin system (RAS) blockers may promote COVID-19 by increasing ACE2, which severe acute respiratory syndrome coronavirus 2 uses to enter host cells, along with the host protease TMPRSS2. Taking a reverse translational approach and by combining in situ hybridization, primary cell isolation, immunoblotting, quantitative RT-PCR, and liquid chromatography-tandem mass spectrometry, we studied lung and kidney ACE2 and TMPRSS2 in diabetic mice mimicking host factors linked to severe COVID-19. In healthy young mice, neither the ACE inhibitor ramipril nor the AT1 receptor blocker telmisartan affected lung or kidney ACE2 or TMPRSS2, except for a small increase in kidney ACE2 protein with ramipril. In contrast, mice with comorbid diabetes (aging, high-fat diet, and streptozotocin-induced diabetes) had heightened lung ACE2 and TMPRSS2 protein levels and increased lung ACE2 activity. None of these parameters were affected by RAS blockade. ACE2 was similarly upregulated in the kidneys of mice with comorbid diabetes compared with aged controls, whereas TMPRSS2 (primarily distal nephron) was highest in telmisartan-treated animals. Upregulation of lung ACE2 activity in comorbid diabetes may contribute to an increased risk of severe COVID-19. This upregulation is driven by comorbidity and not by RAS blockade.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , Diabetes Mellitus, Experimental/metabolism , Diet, High-Fat , Kidney/metabolism , Lung/metabolism , Serine Endopeptidases/genetics , Age Factors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme 2/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , COVID-19 , Immunoblotting , In Situ Hybridization , Kidney/drug effects , Lung/drug effects , Male , Mice , Ramipril/pharmacology , Receptors, Coronavirus/drug effects , Receptors, Coronavirus/genetics , Receptors, Coronavirus/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Serine Endopeptidases/drug effects , Serine Endopeptidases/metabolism , Telmisartan/pharmacology
15.
Circulation ; 120(14): 1401-14, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19770394

ABSTRACT

BACKGROUND: The innate immune system greatly contributes to the inflammatory process after myocardial infarction (MI). Interleukin-1 receptor-associated kinase-4 (IRAK-4), downstream of Toll/interleukin-1 receptor signaling, has an essential role in regulating the innate immune response. The present study was designed to determine the mechanism by which IRAK-4 is responsible for the cardiac inflammatory process, which consequently affects left ventricular remodeling after MI. METHODS AND RESULTS: Experimental MI was created in IRAK-4(-/-) and wild-type mice by left coronary ligation. Mice with a targeted deletion of IRAK-4 had an improved survival rate at 4 weeks after MI. IRAK-4(-/-) mice also demonstrated attenuated cardiac dilation and decreased inflammation in the infarcted myocardium, which was associated with less proinflammatory and Th1 cytokine expression mediated by suppression of nuclear factor-kappaB and c-Jun N-terminal kinase activation. IRAK-4(-/-) mice had fewer infiltrations of CD45+ leukocytes and CD11c+ dendritic cells, inhibition of apoptosis, and reduced fibrosis and nitric oxide production. Cardiac dendritic cells in IRAK-4(-/-) mice were relatively immature or functionally naïve after MI in that they demonstrated less cytokine and costimulatory molecule gene expression. Furthermore, IRAK-4(-/-) dendritic cells have less mobilization capacity. Transfer of wild type-derived bone marrow dendritic cells into IRAK-4(-/-) mice for functional dendritic cell reconstitution negated the survival advantage and reduced the cardiac dilation observed with IRAK-4(-/-) mice at 28 days after MI. CONCLUSIONS: Deletion of IRAK-4 has favorable effects on survival and left ventricular remodeling after MI through modification of the host inflammatory process by blunting the detrimental bone marrow dendritic cells mobilization after myocardial ischemia.


Subject(s)
Bone Marrow Cells/physiology , Dendritic Cells/physiology , Interleukin-1 Receptor-Associated Kinases/physiology , Myocardial Infarction/physiopathology , Ventricular Remodeling/physiology , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Crosses, Genetic , Dendritic Cells/immunology , Disease Models, Animal , Gene Deletion , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Macrophages/immunology , Mice , Mice, Knockout , Myocardial Infarction/immunology , Myocardial Infarction/mortality , Neutrophils/immunology , Polymerase Chain Reaction , Survival Rate , T-Lymphocytes/immunology
16.
Diabetes ; 67(12): 2668-2681, 2018 12.
Article in English | MEDLINE | ID: mdl-30213824

ABSTRACT

The posttranslational histone modifications that epigenetically affect gene transcription extend beyond conventionally studied methylation and acetylation patterns. By examining the means by which podocytes influence the glomerular endothelial phenotype, we identified a role for phosphorylation of histone H3 on serine residue 10 (phospho-histone H3Ser10) in mediating endothelial activation in diabetes. Culture media conditioned by podocytes exposed to high glucose caused glomerular endothelial vascular cell adhesion protein 1 (VCAM-1) upregulation and was enriched for the chemokine CCL2. A neutralizing anti-CCL2 antibody prevented VCAM-1 upregulation in cultured glomerular endothelial cells, and knockout of the CCL2 receptor CCR2 diminished glomerular VCAM-1 upregulation in diabetic mice. CCL2/CCR2 signaling induced glomerular endothelial VCAM-1 upregulation through a pathway regulated by p38 mitogen-activated protein kinase, mitogen- and stress-activated protein kinases 1/2 (MSK1/2), and phosphorylation of H3Ser10, whereas MSK1/2 inhibition decreased H3Ser10 phosphorylation at the VCAM1 promoter. Finally, increased phospho-histone H3Ser10 levels were observed in the kidneys of diabetic endothelial nitric oxide synthase knockout mice and in the glomeruli of humans with diabetic kidney disease. These findings demonstrate the influence that histone protein phosphorylation may have on gene activation in diabetic kidney disease. Histone protein phosphorylation should be borne in mind when considering epigenetic targets amenable to therapeutic manipulation in diabetes.


Subject(s)
Diabetic Nephropathies/metabolism , Endothelium, Vascular/metabolism , Histones/metabolism , Signal Transduction/physiology , Animals , Endothelial Cells/metabolism , Humans , Kidney Glomerulus/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Podocytes/metabolism , Promoter Regions, Genetic , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
17.
Front Pharmacol ; 9: 34, 2018.
Article in English | MEDLINE | ID: mdl-29449811

ABSTRACT

To contend with the deleterious effects of accumulating misfolded protein aggregates or damaged organelles cells rely on a system of quality control processes, among them the autophagy-lysosome pathway. This pathway is itself controlled by a master regulator transcription factor termed transcription factor EB (TFEB). When TFEB localizes to the cell nucleus it promotes the expression of a number of genes involved in protein clearance. Here, we set out to determine (1) whether TFEB expression is altered in chronic kidney disease (CKD); (2) whether inhibition of the cytosolic deacetylase histone deacetylase 6 (HDAC6) affects TFEB acetylation and nuclear localization; and (3) whether HDAC6 inhibition, in turn, alters the natural history of experimental CKD. TFEB mRNA and protein levels were observed to be diminished in the kidneys of humans with diabetic kidney disease, accompanied by accumulation of the protein aggregate adaptor protein p62 in tubule epithelial cells. In cultured NRK-52E cells, HDAC6 inhibition with the small molecule inhibitor Tubastatin A acetylated TFEB, increasing TFEB localization to the nucleus and attenuating cell death. In a rat model of CKD, Tubastatin A prevented the accumulation of misfolded protein aggregates in tubule epithelial cells, attenuated proteinuria progression, limited tubule cell death and diminished tubulointerstitial collagenous matrix deposition. These findings point to the common occurrence of dysregulated quality control processes in CKD and they suggest that TFEB downregulation may contribute to tubule injury in CKD. They also identify a regulatory relationship between HDAC6 and TFEB. HDAC6 inhibitors and TFEB activators both warrant further investigation as treatments for CKD.

18.
J Clin Invest ; 128(1): 483-499, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29227285

ABSTRACT

Histone protein modifications control fate determination during normal development and dedifferentiation during disease. Here, we set out to determine the extent to which dynamic changes to histones affect the differentiated phenotype of ordinarily quiescent adult glomerular podocytes. To do this, we examined the consequences of shifting the balance of the repressive histone H3 lysine 27 trimethylation (H3K27me3) mark in podocytes. Adriamycin nephrotoxicity and subtotal nephrectomy (SNx) studies indicated that deletion of the histone methylating enzyme EZH2 from podocytes decreased H3K27me3 levels and sensitized mice to glomerular disease. H3K27me3 was enriched at the promoter region of the Notch ligand Jag1 in podocytes, and derepression of Jag1 by EZH2 inhibition or knockdown facilitated podocyte dedifferentiation. Conversely, inhibition of the Jumonji C domain-containing demethylases Jmjd3 and UTX increased the H3K27me3 content of podocytes and attenuated glomerular disease in adriamycin nephrotoxicity, SNx, and diabetes. Podocytes in glomeruli from humans with focal segmental glomerulosclerosis or diabetic nephropathy exhibited diminished H3K27me3 and heightened UTX content. Analogous to human disease, inhibition of Jmjd3 and UTX abated nephropathy progression in mice with established glomerular injury and reduced H3K27me3 levels. Together, these findings indicate that ostensibly stable chromatin modifications can be dynamically regulated in quiescent cells and that epigenetic reprogramming can improve outcomes in glomerular disease by repressing the reactivation of developmental pathways.


Subject(s)
Diabetic Nephropathies/metabolism , Histones/metabolism , Podocytes/metabolism , Animals , Diabetic Nephropathies/pathology , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Histone Demethylases/metabolism , Humans , Jagged-1 Protein/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Methylation , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Proteins/metabolism , Podocytes/pathology
19.
Sci Rep ; 7(1): 3442, 2017 06 13.
Article in English | MEDLINE | ID: mdl-28611444

ABSTRACT

The therapeutic targeting of prostanoid subtype receptors may slow the development of chronic kidney disease (CKD) through mechanisms that are distinct from those of upstream COX inhibition. Here, employing multiple experimental models of CKD, we studied the effects of inhibition of the EP4 receptor, one of four receptor subtypes for the prostanoid prostaglandin E2. In streptozotocin-diabetic endothelial nitric oxide synthase knockout mice, EP4 inhibition attenuated the development of albuminuria, whereas the COX inhibitor indomethacin did not. In Type 2 diabetic db/db mice, EP4 inhibition lowered albuminuria to a level comparable with that of the ACE inhibitor captopril. However, unlike captopril, EP4 inhibition had no effect on blood pressure or hyperfiltration although it did attenuate mesangial matrix accumulation. Indicating a glucose-independent mechanism of action, EP4 inhibition also attenuated proteinuria development and glomerular scarring in non-diabetic rats subjected to surgical renal mass ablation. Finally, in vitro, EP4 inhibition prevented transforming growth factor-ß1 induced dedifferentiation of glomerular podocytes. In rodent models of diabetic and non-diabetic CKD, EP4 inhibition attenuated renal injury through mechanisms that were distinct from either broadspectrum COX inhibition or "standard of care" renin angiotensin system blockade. EP4 inhibition may represent a viable repurposing opportunity for the treatment of CKD.


Subject(s)
Diabetic Nephropathies/drug therapy , Naphthalenes/pharmacology , Phenylbutyrates/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Renal Insufficiency, Chronic/drug therapy , Animals , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Naphthalenes/therapeutic use , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phenylbutyrates/therapeutic use , Podocytes/drug effects , Podocytes/metabolism , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/metabolism
20.
Circulation ; 112(15): 2276-85, 2005 Oct 11.
Article in English | MEDLINE | ID: mdl-16216974

ABSTRACT

BACKGROUND: Myeloid differentiation factor (MyD)-88 is a key adaptor protein that plays a major role in the innate immune pathway. How MyD88 may regulate host response in inflammatory heart disease is unknown. METHODS AND RESULTS: We found that the cardiac protein level of MyD88 was significantly increased in the hearts of wild-type mice after exposure to Coxsackievirus B3 (CVB3). MyD88(-/-) mice showed a dramatic higher survival rate (86%) in contrast to the low survival (35%) in the MyD88(+/+) mice after CVB3 infection (P<0.0001). Pathological examination showed a significant decrease of cardiac and pancreatic inflammation in the MyD88(-/-) mice. Viral concentrations in the hearts were significantly decreased in the MyD88(-/-) mice. Cardiac mRNA levels for interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma, and IL-18 were significantly decreased in the MyD88(-/-) mice. Similarly, serum levels of T-helper 1 cytokines were significantly decreased in the MyD88(-/-) mice. In contrast, cardiac protein levels of the activated interferon regulatory factor (IRF)-3 and IFN-beta were significantly increased in the MyD88(-/-) mice but not other usual upstream signals to IRF-3. The cardiac expression of coxsackie-adenoviral receptor and p56(lck) were also significantly decreased. CONCLUSIONS: MyD88 appears to be a key contributor to cardiac inflammation, mediating cytokine production and T-helper-1/2 cytokine balance, increasing coxsackie-adenoviral receptor and p56(lck) expression and viral titers after CVB3 exposure. Absence of MyD88 confers host protection possibly through novel direct activation of IRF-3 and IFN-beta.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Enterovirus B, Human , Enterovirus Infections/complications , Interferon Type I/biosynthesis , Myocarditis/virology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , DNA Primers , Disease Models, Animal , Enterovirus Infections/immunology , HeLa Cells , Heart/virology , Humans , Immunity, Innate , Inflammation/immunology , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88 , Myocarditis/immunology , Myocarditis/physiopathology , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL