Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(13): e23780, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38948938

ABSTRACT

Aerobic training (AT), an effective form of cardiac rehabilitation, has been shown to be beneficial for cardiac repair and remodeling after myocardial infarction (MI). The p300/CBP-associated factor (PCAF) is one of the most important lysine acetyltransferases and is involved in various biological processes. However, the role of PCAF in AT and AT-mediated cardiac remodeling post-MI has not been determined. Here, we found that the PCAF protein level was significantly increased after MI, while AT blocked the increase in PCAF. AT markedly improved cardiac remodeling in mice after MI by reducing endoplasmic reticulum stress (ERS). In vivo, similar to AT, pharmacological inhibition of PCAF by Embelin improved cardiac recovery and attenuated ERS in MI mice. Furthermore, we observed that both IGF-1, a simulated exercise environment, and Embelin protected from H2O2-induced cardiomyocyte injury, while PCAF overexpression by viruses or the sirtuin inhibitor nicotinamide eliminated the protective effect of IGF-1 in H9C2 cells. Thus, our data indicate that maintaining low PCAF levels plays an essential role in AT-mediated cardiac protection, and PCAF inhibition represents a promising therapeutic target for attenuating cardiac remodeling after MI.


Subject(s)
Myocardial Infarction , Physical Conditioning, Animal , Ventricular Remodeling , p300-CBP Transcription Factors , Animals , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Mice , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Endoplasmic Reticulum Stress/drug effects
3.
Adv Sci (Weinh) ; 11(18): e2307926, 2024 May.
Article in English | MEDLINE | ID: mdl-38460156

ABSTRACT

Prostanoids are endogenous lipid bioactive mediators that play essential roles in physiological processes such as glucocorticoid secretion. Here, it is found that the thromboxane (Tx)A2 receptor (TP) is highly expressed in the adrenal cortex of mice. Both global and adrenocortical-specific deletion of the TP receptor lead to increased adiposity in mice by elevating corticosterone synthesis. Mechanistically, the TP receptor deletion increases the phosphorylation of steroidogenic acute regulatory protein (StAR) and corticosterone synthesis in adrenal cortical cells by suppressing p-p38-mediated phosphorylation of 14-3-3γ adapter protein at S71. The activation of the p38 in the adrenal cortical cells by forced expression of the MKK6EE gene attenuates hypercortisolism in TP-deficient mice. These observations suggest that the TxA2/TP signaling regulates adrenal corticosterone homeostasis independent of the hypothalamic-pituitary-adrenal axis and the TP receptor may serve as a promising therapeutic target for hypercortisolism.


Subject(s)
Corticosterone , Phosphoproteins , Signal Transduction , Thromboxane A2 , Animals , Mice , Corticosterone/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Thromboxane A2/metabolism , Adrenal Cortex/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , Male , Mice, Inbred C57BL
4.
Redox Biol ; 65: 102825, 2023 09.
Article in English | MEDLINE | ID: mdl-37531930

ABSTRACT

Chemotherapeutic agents, such as doxorubicin (DOX), may cause cardiomyopathy, even life-threatening arrhythmias in cancer patients. Ferroptosis-an iron-dependent oxidative form of programmed necrosis, plays a pivotal role in DOX-induced cardiomyopathy (DIC). Prostaglandins (PGs) are bioactive signaling molecules that profoundly modulate cardiac performance in both physiologic and pathologic conditions. Here, we found that PGE2 production and its E-prostanoid 1 receptor (EP1) expression were upregulated in erastin (a ferroptosis inducer) or DOX-treated cardiomyocytes. EP1 inhibition markedly aggravated erastin or DOX-induced cardiomyocyte ferroptosis, whereas EP1 activation exerted opposite effect. Genetic depletion of EP1 in cardiomyocytes worsens DOX-induced cardiac injury in mice, which was efficiently rescued by the ferroptosis inhibitor Ferrostatin-1 (Fer-1). Mechanistically, EP1 activation protected cardiomyocytes from DOX-induced ferroptosis by promoting nuclear factor erythroid 2-related factor 2 (Nrf2)-driven anti-oxidative gene expression, such as glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11). EP1 was coupled with Gαq to elicit intracellular Ca2+ flux and activate the PKC/Nrf2 cascade in ferroptotic cardiomyocytes. EP1 activation also prevents DOX-induced ferroptosis in human cardiomyocytes. Thus, PGE2/EP1 axis protects cardiomyocytes from DOX-induced ferroptosis by activating PKC/Nrf2 signaling and activation of EP1 may represent an attractive strategy for DIC prevention and treatment.


Subject(s)
Ferroptosis , Animals , Humans , Mice , Apoptosis , Dinoprostone , Doxorubicin/adverse effects , Myocytes, Cardiac , NF-E2-Related Factor 2/genetics
5.
Biochem Biophys Res Commun ; 624: 8-15, 2022 10 08.
Article in English | MEDLINE | ID: mdl-35932581

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of central vision loss in the elderly. Oxidative stress-induced retinal pigment epithelium (RPE) cell apoptosis is a crucial pathogenic hallmark in AMD. Chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2), a prostaglandin (PG) D2 receptor, has been implicated in various pathophysiological events, especially inflammation and stress-induced cell apoptosis. However, its specific role in AMD is not fully understood. Here we studied the effect of CRTH2 on AMD. Our results showed that when stimulated by H2O2, CRTH2 mRNA expression in cells tended to increase. Flow cytometry revealed that the CRTH2 inhibitor could protect the RPE from apoptosis. After NaIO3 injection, a larger area of retinal degeneration was observed in wild-type mice than in CRTH2-/- mice. Optical coherence tomography (OCT) and Hematoxylin and Eosin (H&E) staining of retinal sections showed that sodium iodate-induced loss of photoreceptor cells was reduced in CRTH2-/- mice after treatment; TUNEL-positive cells were mostly found in the outer nuclear layer. In the control group, NaIO3 stimulation increased the number of TUNEL-positive cells, whereas the percentage of TUNEL-positive cells was significantly lower in CRTH2-/- mice. Similarly, the CRTH2 receptor inhibitor CAY10471 similarly inhibited sodium iodate-induced retinal damage. Our results suggest that targeting CRTH2 is a promising therapeutic strategy for the treatment of progressive retinal degeneration in AMD.


Subject(s)
Macular Degeneration , Retinal Degeneration , Animals , Disease Models, Animal , Hydrogen Peroxide/metabolism , Macular Degeneration/genetics , Mice , Oxidative Stress , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism
6.
Cells ; 11(15)2022 07 30.
Article in English | MEDLINE | ID: mdl-35954191

ABSTRACT

Cardiac fibrosis is a common pathophysiologic process associated with numerous cardiovascular diseases, resulting in cardiac dysfunction. Cardiac fibroblasts (CFs) play an important role in the production of the extracellular matrix and are the essential cell type in a quiescent state in a healthy heart. In response to diverse pathologic stress and environmental stress, resident CFs convert to activated fibroblasts, referred to as myofibroblasts, which produce more extracellular matrix, contributing to cardiac fibrosis. Although multiple molecular mechanisms are implicated in CFs activation and cardiac fibrosis, there is increasing evidence that epigenetic regulation plays a key role in this process. Epigenetics is a rapidly growing field in biology, and provides a modulated link between pathological stimuli and gene expression profiles, ultimately leading to corresponding pathological changes. Epigenetic modifications are mainly composed of three main categories: DNA methylation, histone modifications, and non-coding RNAs. This review focuses on recent advances regarding epigenetic regulation in cardiac fibrosis and highlights the effects of epigenetic modifications on CFs activation. Finally, we provide some perspectives and prospects for the study of epigenetic modifications and cardiac fibrosis.


Subject(s)
Epigenesis, Genetic , Fibroblasts/metabolism , Fibrosis , Heart , Humans
7.
EMBO J ; 41(16): e110439, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35781818

ABSTRACT

Brown adipose tissue (BAT) functions as a thermogenic organ and is negatively associated with cardiometabolic diseases. N6 -methyladenosine (m6 A) modulation regulates the fate of stem cells. Here, we show that the prostaglandin E2 (PGE2 )-E-prostanoid receptor 3 (EP3) axis was activated during mouse interscapular BAT development. Disruption of EP3 impaired the browning process during adipocyte differentiation from pre-adipocytes. Brown adipocyte-specific depletion of EP3 compromised interscapular BAT formation and aggravated high-fat diet-induced obesity and insulin resistance in vivo. Mechanistically, activation of EP3 stabilized the Zfp410 mRNA via WTAP-mediated m6 A modification, while knockdown of Zfp410 abolished the EP3-induced enhancement of brown adipogenesis. EP3 prevented ubiquitin-mediated degradation of WTAP by eliminating PKA-mediated ERK1/2 inhibition during brown adipocyte differentiation. Ablation of WTAP in brown adipocytes abrogated the protective effect of EP3 overexpression in high-fat diet-fed mice. Inhibition of EP3 also retarded human embryonic stem cell differentiation into mature brown adipocytes by reducing the WTAP levels. Thus, a conserved PGE2 -EP3 axis promotes BAT development by stabilizing WTAP/Zfp410 signaling in a PKA/ERK1/2-dependent manner.


Subject(s)
Adipose Tissue, Brown , Dinoprostone , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Animals , Cell Cycle Proteins/metabolism , Dinoprostone/metabolism , Humans , Methyltransferases/metabolism , Mice , RNA/metabolism , RNA Splicing Factors/metabolism , Receptors, Prostaglandin E, EP3 Subtype , Thermogenesis
8.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35420633

ABSTRACT

Natural killer (NK) cells exhibit antifibrotic properties in liver fibrosis (LF) by suppressing activated hepatic stellate cell (HSC) populations. Prostaglandin E2 (PGE2) plays a dual role in innate and adaptive immunity. Here, we found that E-prostanoid 3 receptor (EP3) was markedly downregulated in NK cells from liver fibrosis mice and patients with liver cirrhosis. NK cell-specific deletion of EP3 aggravated hepatic fibrogenesis in mouse models of LF. Loss of EP3 selectively reduced the cytotoxicity of the CD27+CD11b+ double positive (DP) NK subset against activated HSCs. Mechanistically, deletion of EP3 impaired the adhesion and cytotoxicity of DP NK cells toward HSCs through modulation of Itga4-VCAM1 binding. EP3 upregulated Itga4 expression in NK cells through promoting Spic nuclear translocation via PKC-mediated phosphorylation of Spic at T191. Activation of EP3 by sulprostone alleviated CCL4-induced liver fibrosis in mice. Thus, EP3 is required for adhesion and cytotoxicity of NK cells toward HSCs and may serve as a therapeutic target for the management of LF.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Animals , Disease Models, Animal , Hepatic Stellate Cells/metabolism , Humans , Killer Cells, Natural , Liver Cirrhosis/metabolism , Mice
9.
Sheng Li Xue Bao ; 73(4): 559-570, 2021 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-34405212

ABSTRACT

Prostaglandins are a class of poly-unsaturated fatty acids-derived bioactive lipids with important physiological function by binding to specific receptors. Prostaglandin receptors lack specific antibodies, which greatly impedes the research on our understanding of the signaling of prostaglandins. The aim of this study was to identify nine mouse lines with amino terminal (-NH2, -N) HA-tagged prostaglandin receptors by using the combination of artificial sperm and CRISPR-Cas9 technology. The guide RNA expression plasmid and labeled targeting vector plasmids were transferred into "artificial sperm cells". The "artificial sperm cells" containing labeled proteins were selected and injected into mouse oocytes, and implanted into pseudopregnant mice to obtain labeled mice. The genomic DNA of the prostaglandin receptor tagged mice was extracted, and the genotypes of mice were detected by PCR method. We also isolated mouse peritoneal macrophages to verify the protein expression of HA-labeled prostaglandin receptor by Western blot. Specific DNA bands were amplified in prostaglandin receptor labeled mice, and specific HA protein bands were detected in macrophage proteins, which was not detected in wild type mice. In summary, we successfully constructed 9 mouse lines with HA-tagged prostaglandin receptors, providing a powerful tool for further study of the pathophysiological functions of prostaglandin signaling both in vivo and in vitro.


Subject(s)
RNA, Guide, Kinetoplastida , Receptors, Prostaglandin , Animals , Mice , Oocytes , Plasmids
10.
EMBO J ; 40(16): e107403, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34223653

ABSTRACT

Excessive deposition of extracellular matrix, mainly collagen protein, is the hallmark of organ fibrosis. The molecular mechanisms regulating fibrotic protein biosynthesis are unclear. Here, we find that chemoattractant receptor homologous molecule expressed on TH2 cells (CRTH2), a plasma membrane receptor for prostaglandin D2, is trafficked to the endoplasmic reticulum (ER) membrane in fibroblasts in a caveolin-1-dependent manner. ER-anchored CRTH2 binds the collagen mRNA recognition motif of La ribonucleoprotein domain family member 6 (LARP6) and promotes the degradation of collagen mRNA in these cells. In line, CRTH2 deficiency increases collagen biosynthesis in fibroblasts and exacerbates injury-induced organ fibrosis in mice, which can be rescued by LARP6 depletion. Administration of CRTH2 N-terminal peptide reduces collagen production by binding to LARP6. Similar to CRTH2, bumetanide binds the LARP6 mRNA recognition motif, suppresses collagen biosynthesis, and alleviates bleomycin-triggered pulmonary fibrosis in vivo. These findings reveal a novel anti-fibrotic function of CRTH2 in the ER membrane via the interaction with LARP6, which may represent a therapeutic target for fibrotic diseases.


Subject(s)
Autoantigens/metabolism , Collagen/antagonists & inhibitors , Liver Cirrhosis/prevention & control , Pulmonary Fibrosis/prevention & control , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Ribonucleoproteins/metabolism , Animals , Bleomycin , Carbon Tetrachloride , Cells, Cultured , Collagen/biosynthesis , Collagen/genetics , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Intracellular Membranes/metabolism , Isoproterenol , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Lung/metabolism , Lung/pathology , Male , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Protein Binding , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , SS-B Antigen
11.
Biochem Biophys Res Commun ; 561: 7-13, 2021 07 05.
Article in English | MEDLINE | ID: mdl-33992835

ABSTRACT

Doxorubicin (DOX) is a broad-spectrum antineoplastic drug; however, its serious cardiotoxic side effects in inflammatory responses limit its use in clinical applications. Dopamine D1 receptor (DRD1), a G protein-coupled receptor, is crucial for the development and function of the nervous system; additionally, it also play a role in immune regulation. However, the specific role of DRD1 in DOX-induced cardiac inflammation has not yet been clarified. Here, we discovered that DRD1 expression was induced by DOX treatment in H9C2 cardiomyocytes. DRD1 activation by A-68930, a DRD1-specific agonist, decreased DOX-induced nucleotide-binding domain-like receptor protein 3 (NLRP3) expression, caspase-1 activation, and IL-1ß maturation in H9C2 cells. Expression of the cytokines IL-1ß and IL-18 in the supernatants was also inhibited by A-68930 treatment. DRD1 knockdown, using siRNA, abolished the effects of A-68930 on the DOX-induced NLRP3 inflammasome. Furthermore, we found that DRD1 signaling downregulated the NLRP3 inflammasome in H9C2 cells through cyclic adenosine monophosphate (cAMP). Moreover, application of A-68930 to activate DRD1 reduced cardiac injury and fibrosis in a DOX-treated mouse model by suppressing the NLRP3 inflammasome in the heart. These findings indicate that DRD1 signaling may protect against DOX-induced cardiac injury by inhibiting the NLRP3 inflammasome-mediated inflammation.


Subject(s)
Cardiotoxicity/prevention & control , Chromans/pharmacology , Doxorubicin/toxicity , Inflammasomes/antagonists & inhibitors , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Receptors, Dopamine D1/agonists , Animals , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Cells, Cultured , Cytokines/metabolism , Dopamine Agonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Receptors, Dopamine D1/metabolism , Signal Transduction , Topoisomerase II Inhibitors/toxicity
12.
FEBS Open Bio ; 11(7): 2033-2040, 2021 07.
Article in English | MEDLINE | ID: mdl-34056872

ABSTRACT

Cardiac fibrosis is the final event of heart failure and is associated with almost all forms of cardiovascular disease. Cardiac fibroblasts (CFs), a major cell type in the heart, are responsible for regulating normal myocardial function and maintaining extracellular matrix homeostasis in adverse myocardial remodeling. In this study, we found that C188-9, a small-molecule inhibitor of signal transducer and activator of transcription 3 (STAT3), exhibited an antifibrotic function, both in vitro and in vivo. C188-9 decreased transforming growth factor-ß1-induced CF activation and fibrotic gene expression. Moreover, C188-9 treatment alleviated heart injury and cardiac fibrosis in an isoproterenol-induced mouse model by suppressing STAT3 phosphorylation and activation. These findings may help us better understand the role of C188-9 in cardiac fibrosis and facilitate the development of new treatments for cardiac fibrosis and other cardiovascular diseases.


Subject(s)
Fibroblasts , Transforming Growth Factor beta1 , Animals , Fibroblasts/metabolism , Fibrosis , Isoproterenol/metabolism , Isoproterenol/pharmacology , Mice , Myocardium , Transforming Growth Factor beta1/metabolism
14.
Sci Adv ; 6(42)2020 10.
Article in English | MEDLINE | ID: mdl-33055159

ABSTRACT

Cardiovascular dysfunction is one of the most common complications of long-term cancer treatment. Growing evidence has shown that antineoplastic drugs can increase cardiovascular risk during cancer therapy, seriously affecting patient survival. However, little is known about the genetic factors associated with the cardiovascular risk of antineoplastic drugs. We established a compendium of genetic evidence that supports cardiovascular risk induced by antineoplastic drugs. Most of this genetic evidence is attributed to causal alleles altering the expression of cardiovascular disease genes. We found that antineoplastic drugs predicted to induce cardiovascular risk are significantly enriched in drugs associated with cardiovascular adverse reactions, including many first-line cancer treatments. Functional experiments validated that retinoid X receptor agonists can reduce triglyceride lipolysis, thus modulating cardiovascular risk. Our results establish a link between the causal allele of cardiovascular disease genes and the direction of pharmacological modulation, which could facilitate cancer drug discovery and clinical trial design.


Subject(s)
Antineoplastic Agents , Cardiovascular Diseases , Neoplasms , Antineoplastic Agents/adverse effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/genetics , Heart Disease Risk Factors , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Risk Factors
15.
FASEB J ; 34(11): 14892-14904, 2020 11.
Article in English | MEDLINE | ID: mdl-32939891

ABSTRACT

Renal fibrosis is a common pathological hallmark of chronic kidney disease (CKD). Renal sympathetic nerve activity is elevated in patients and experimental animals with CKD and contributes to renal interstitial fibrosis in obstructive nephropathy. However, the mechanisms underlying sympathetic overactivation in renal fibrosis remain unknown. Norepinephrine (NE), the main sympathetic neurotransmitter, was found to promote TGF-ß1-induced epithelial-mesenchymal transition (EMT) and fibrotic gene expression in the human renal proximal epithelial cell line HK-2. Using both genetic and pharmacological approaches, we identified that NE binds Gαq-coupled α1-adrenoceptor (α1-AR) to enhance EMT of HK-2 cells by activating p38/Smad3 signaling. Inhibition of p38 diminished the NE-exaggerated EMT process and increased the fibrotic gene expression in TGF-ß1-treated HK-2 cells. Moreover, the pharmacological blockade of α1-AR reduced the kidney injury and renal fibrosis in a unilateral ureteral obstruction mouse model by suppressing EMT in the kidneys. Thus, sympathetic overactivation facilitates EMT of renal epithelial cells and fibrosis via the α1-AR/p38/Smad3 signaling pathway, and α1-AR inhibition may be a promising approach toward treating renal fibrosis.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Renal Insufficiency, Chronic/metabolism , Tamsulosin/pharmacology , Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Adrenergic alpha-Agonists/pharmacology , Animals , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Norepinephrine/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/etiology , Smad3 Protein/metabolism , Tamsulosin/therapeutic use , Transforming Growth Factor beta/pharmacology , Urethral Obstruction/complications , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Circulation ; 141(8): 655-666, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31893939

ABSTRACT

BACKGROUND: Blood pressure often rises with aging, but exact mechanisms are still not completely understood. With aging, the level of proinflammatory cytokines increases in T lymphocytes. Prostaglandin D2, a proresolution mediator, suppresses Type 1 T helper (Th1) cytokines through D-prostanoid receptor 1 (DP1). In this study, we aimed to investigate the role of the prostaglandin D2/DP1 axis in T cells on age-related hypertension. METHODS: To clarify the physiological and pathophysiological roles of DP1 in T cells with aging, peripheral blood samples were collected from young and older male participants, and CD4+ T cells were sorted for gene expression, prostaglandin production, and Western blot assays. Mice blood pressure was quantified by invasive telemetric monitor. RESULTS: The prostaglandin D2/DP1 axis was downregulated in CD4+ T cells from older humans and aged mice. DP1 deletion in CD4+ T cells augmented age-related hypertension in aged male mice by enhancing Th1 cytokine secretion, vascular remodeling, CD4+ T cells infiltration, and superoxide production in vasculature and kidneys. Conversely, forced expression of exogenous DP1 in T cells retarded age-associated hypertension in mice by reducing Th1 cytokine secretion. Tumor necrosis factor α neutralization or interferon γ deletion ameliorated the age-related hypertension in DP1 deletion in CD4+ T cells mice. Mechanistically, DP1 inhibited Th1 activity via the PKA (protein kinase A)/p-Sp1 (phosphorylated specificity protein 1)/neural precursor cell expressed developmentally downregulated 4-like (NEDD4L) pathway-mediated T-box-expressed-in-T-cells (T-bet) ubiquitination. T-bet deletion or forced NEDD4L expression in CD4+ T cells attenuated age-related hypertension in CD4+ T cell-specific DP1-deficient mice. DP1 receptor activation by BW245C prevented age-associated blood pressure elevation and reduced vascular/renal superoxide production in male mice. CONCLUSIONS: The prostaglandin D2/DP1 axis suppresses age-related Th1 activation and subsequent hypertensive response in male mice through increase of NEDD4L-mediated T-bet degradation by ubiquitination. Therefore, the T cell DP1 receptor may be an attractive therapeutic target for age-related hypertension.


Subject(s)
Aging , CD4-Positive T-Lymphocytes/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Receptors, Prostaglandin/metabolism , T-Box Domain Proteins/metabolism , Aged , Animals , Antihypertensive Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/metabolism , Humans , Hypertension/drug therapy , Hypertension/pathology , Mice , Mice, Inbred C57BL , Prostaglandin D2/metabolism , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/deficiency , Receptors, Prostaglandin/genetics , Signal Transduction , Sp1 Transcription Factor/metabolism , Superoxides/metabolism , Th1 Cells/metabolism , Ubiquitination
17.
J Exp Med ; 215(8): 2175-2195, 2018 08 06.
Article in English | MEDLINE | ID: mdl-29970474

ABSTRACT

Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by progressive pulmonary artery (PA) remodeling. T helper 2 cell (Th2) immune response is involved in PA remodeling during PAH progression. Here, we found that CRTH2 (chemoattractant receptor homologous molecule expressed on Th2 cell) expression was up-regulated in circulating CD3+CD4+ T cells in patients with idiopathic PAH and in rodent PAH models. CRTH2 disruption dramatically ameliorated PA remodeling and pulmonary hypertension in different PAH mouse models. CRTH2 deficiency suppressed Th2 activation, including IL-4 and IL-13 secretion. Both CRTH2+/+ bone marrow reconstitution and CRTH2+/+ CD4+ T cell adoptive transfer deteriorated hypoxia + ovalbumin-induced PAH in CRTH2-/- mice, which was reversed by dual neutralization of IL-4 and IL-13. CRTH2 inhibition alleviated established PAH in mice by repressing Th2 activity. In culture, CRTH2 activation in Th2 cells promoted pulmonary arterial smooth muscle cell proliferation through activation of STAT6. These results demonstrate the critical role of CRTH2-mediated Th2 response in PAH pathogenesis and highlight the CRTH2 receptor as a potential therapeutic target for PAH.


Subject(s)
Hypertension, Pulmonary/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Th2 Cells/immunology , Adoptive Transfer , Adult , Animals , Antibodies/pharmacology , Blood Pressure/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Proliferation/drug effects , Chimera , Chronic Disease , Disease Models, Animal , Female , Gene Deletion , Humans , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Immunity/drug effects , Indoles , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Mice , Ovalbumin , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Pyrroles , Receptors, Immunologic/deficiency , Receptors, Prostaglandin/deficiency , STAT6 Transcription Factor/metabolism , Up-Regulation/drug effects
18.
Diabetes ; 67(9): 1748-1760, 2018 09.
Article in English | MEDLINE | ID: mdl-29773555

ABSTRACT

Gluconeogenesis is drastically increased in patients with type 2 diabetes and accounts for increased fasting plasma glucose concentrations. Circulating levels of prostaglandin (PG) F2α are also markedly elevated in diabetes; however, whether and how PGF2α regulates hepatic glucose metabolism remain unknown. Here, we demonstrated that PGF2α receptor (F-prostanoid receptor [FP]) was upregulated in the livers of mice upon fasting- and diabetic stress. Hepatic deletion of the FP receptor suppressed fasting-induced hepatic gluconeogenesis, whereas FP overexpression enhanced hepatic gluconeogenesis in mice. FP activation promoted the expression of gluconeogenic enzymes (PEPCK and glucose-6-phosphatase) in hepatocytes in a FOXO1-dependent manner. Additionally, FP coupled with Gq in hepatocytes to elicit Ca2+ release, which activated Ca2+/calmodulin-activated protein kinase IIγ (CaMKIIγ) to increase FOXO1 phosphorylation and subsequently accelerate its nuclear translocation. Blockage of p38 disrupted CaMKIIγ-induced FOXO1 nuclear translocation and abrogated FP-mediated hepatic gluconeogenesis in mice. Moreover, knockdown of hepatic FP receptor improved insulin sensitivity and glucose homeostasis in ob/ob mice. FP-mediated hepatic gluconeogenesis via the CaMKIIγ/p38/FOXO1 signaling pathway, indicating that the FP receptor might be a promising therapeutic target for type 2 diabetes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dinoprost/metabolism , Forkhead Box Protein O1/metabolism , Gluconeogenesis , Liver/metabolism , Obesity/metabolism , Receptors, Prostaglandin/agonists , Active Transport, Cell Nucleus/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Crosses, Genetic , Diet, High-Fat/adverse effects , Fasting/metabolism , Forkhead Box Protein O1/agonists , Forkhead Box Protein O1/genetics , Gene Expression Regulation/drug effects , Gluconeogenesis/drug effects , Humans , Insulin Resistance , Liver/cytology , Liver/drug effects , Liver/pathology , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Obesity/etiology , Obesity/pathology , Protein Kinase Inhibitors/pharmacology , RNA Interference , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
EMBO Mol Med ; 10(3)2018 03.
Article in English | MEDLINE | ID: mdl-29335338

ABSTRACT

Apoptotic death of cardiac myocytes is associated with ischemic heart disease and chemotherapy-induced cardiomyopathy. Chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) is highly expressed in the heart. However, its specific role in ischemic cardiomyopathy is not fully understood. Here, we demonstrated that CRTH2 disruption markedly improved cardiac recovery in mice postmyocardial infarction and doxorubicin challenge by suppressing cardiomyocyte apoptosis. Mechanistically, CRTH2 activation specifically facilitated endoplasmic reticulum (ER) stress-induced cardiomyocyte apoptosis via caspase-12-dependent pathway. Blockage of m-calpain prevented CRTH2-mediated cardiomyocyte apoptosis under ER stress by suppressing caspase-12 activity. CRTH2 was coupled with Gαq to elicit intracellular Ca2+ flux and activated m-calpain/caspase-12 cascade in cardiomyocytes. Knockdown of caspase-4, an alternative to caspase-12 in humans, markedly alleviated CRHT2 activation-induced apoptosis in human cardiomyocyte response to anoxia. Our findings revealed an unexpected role of CRTH2 in promoting ER stress-induced cardiomyocyte apoptosis, suggesting that CRTH2 inhibition has therapeutic potential for ischemic cardiomyopathy.


Subject(s)
Apoptosis , Calpain/metabolism , Endoplasmic Reticulum Stress , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Animals , Apoptosis/drug effects , Bone Marrow/pathology , Calcium/metabolism , Cardiotonic Agents/pharmacology , Caspase 12/metabolism , Cell Hypoxia/drug effects , Cellular Reprogramming/genetics , Doxorubicin/pharmacology , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Deletion , Humans , Male , Mice , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Prostaglandin D2/metabolism , Regeneration/drug effects , Tetrazoles/pharmacology
20.
Br J Pharmacol ; 175(8): 1205-1216, 2018 04.
Article in English | MEDLINE | ID: mdl-28925017

ABSTRACT

BACKGROUND AND PURPOSE: An appropriate inflammatory response is necessary for cardiac healing after acute myocardial infarction (MI). Resolvin E1 (RvE1) is an anti-inflammatory and pro-resolution lipid mediator derived from eicosapentaenoic acid. Here we have investigated the effects of RvE1 on the recovery of cardiac function after MI in mice. EXPERIMENTAL APPROACH: Acute MI was induced by surgical ligation of the left anterior descending artery in male C57BL/6 mice. RvE1 (5 ng·g-1 ·day-1 ; i.p.) was given to mice at different times following MI. Cardiac function was monitored by transthoracic echocardiography at days 3, 7 and 14 after MI. Effects of RvE1 on the migration of subpopulations of monocytes/macrophages (Mos/Mps, Ly6Chi and Ly6Clow ) were examined by flow cytometry and transwell assay. KEY RESULTS: RvE1 administration from days 1 to 7 post-MI improved cardiac function, whereas treatment from days 7 to 14 markedly inhibited recovery of cardiac function. Early treatment with RvE1 post-MI suppressed the infiltration of dominant Ly6Chi Mos/Mps and secretion of pro-inflammatory cytokines in injured hearts, which protected cardiomyocytes against apoptosis in the peri-infarct zones. Contrastingly, treatment with RvE1 1 week after MI decreased infiltration of Ly6Clow Mos/Mps and expression of pro-angiogenic factors in cardiac tissue, consequently reducing neovascularization in the peri-infarct zones. Additionally, RvE1 inhibited Mp migration by activating ChemR23 receptors. CONCLUSION AND IMPLICATIONS: Treatment with RvE1 during the initial 7 days after MI facilitated cardiac healing by suppressing pro-inflammatory cytokine secretion, indicating that RvE1 may serve as an early therapeutic agent for acute MI. LINKED ARTICLES: This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.


Subject(s)
Eicosapentaenoic Acid/analogs & derivatives , Myocardial Infarction/drug therapy , Adoptive Transfer , Animals , Cell Movement/drug effects , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/therapeutic use , Fibroblasts/drug effects , Fibroblasts/physiology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/physiology , Male , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/physiology , Monocytes/transplantation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology
SELECTION OF CITATIONS
SEARCH DETAIL