Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Free Radic Biol Med ; 219: 141-152, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636714

ABSTRACT

Pulmonary hypertension (PH) is a devastating disease that lacks effective treatment options and is characterized by severe pulmonary vascular remodeling. Pulmonary arterial endothelial cell (PAEC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension. Canonical transient receptor potential (TRPC) channels, a family of Ca2+-permeable channels, play an important role in various diseases. However, the effect and mechanism of TRPCs on PH development have not been fully elucidated. Among the TRPC family members, TRPC4 expression was markedly upregulated in PAECs from hypoxia combined with SU5416 (HySu)-induced PH mice and monocrotaline (MCT)-treated PH rats, as well as in hypoxia-exposed PAECs, suggesting that TRPC4 in PAECs may participate in the occurrence and development of PH. In this study, we aimed to investigate whether TRPC4 in PAECs has an aggravating effect on PH and elucidate the molecular mechanisms. We observed that hypoxia treatment promoted PAEC apoptosis through a caspase-12/endoplasmic reticulum stress (ERS)-dependent pathway. Knockdown of TRPC4 attenuated hypoxia-induced apoptosis and caspase-3/caspase-12 activity in PAECs. Accordingly, adeno-associated virus (AAV) serotype 6-mediated pulmonary endothelial TRPC4 silencing (AAV6-Tie-shRNA-TRPC4) or TRPC4 antagonist suppressed PH progression as evidenced by reduced right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, PAEC apoptosis and reactive oxygen species (ROS) production. Mechanistically, unbiased RNA sequencing (RNA-seq) suggested that TRPC4 deficiency suppressed the expression of the proapoptotic protein sushi domain containing 2 (Susd2) in hypoxia-exposed mouse PAECs. Moreover, TRPC4 activated hypoxia-induced PAEC apoptosis by promoting Susd2 expression. Therefore, inhibiting TRPC4 ameliorated PAEC apoptosis and hypoxic PH in animals by repressing Susd2 signaling, which may serve as a therapeutic target for the management of PH.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Endothelial Cells , Hypertension, Pulmonary , Hypoxia , TRPC Cation Channels , Animals , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/genetics , Rats , Hypoxia/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Male , Monocrotaline/toxicity , Vascular Remodeling/genetics , Disease Models, Animal , Humans , Signal Transduction , Mice, Inbred C57BL , Rats, Sprague-Dawley , Cells, Cultured , Indoles , Pyrroles
3.
J Colloid Interface Sci ; 652(Pt B): 1925-1936, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37690300

ABSTRACT

Accurate detection and differentiation of multiple anions is still a difficult problem due to their wide variety, structural similarity, and mutual interference. Hence, four rare-earth metal-organic frameworks (RE-MOFs) including Dy-MOFs, Er-MOFs, Tb-MOFs and Y-MOFs are successfully prepared by using TCPP as the ligand and rare-earth ions as the metal center via coordination chelation. It is found that 7 anions can light up their fluorescence. Thus, a high-resolution sensing array based on RE-MOFs nanoprobes is employed to differentiate these anions from intricate analytes in real-time scenarios. The distinctive host-guest response promotes the RE-MOFs nanoprobes to selectively extract the target anions from the complex samples. By taking advantage of the cross-response between RE-MOFs nanoprobes and anions, it allows to create an array for detecting target analytes using pattern recognition. Additionally, RE-MOFs nanoprobes also facilitate the quantitative analysis of these anions (PO43-, H2PO4-, HPO42-, F-, S2-, CO32- and C2O42-). More importantly, the exceptional effectiveness of this method has been demonstrated through various successful applications, including quality monitoring of 8 toothpaste brands, intracellular phosphate imaging, and blood phosphorus detection in mice with vascular calcification. These findings provide robust evidence for the efficacy and reliability of the RE-MOFs nanoprobes array for anion recognition.

5.
Front Cell Dev Biol ; 11: 1132060, 2023.
Article in English | MEDLINE | ID: mdl-37009479

ABSTRACT

Pulmonary hypertension (PH) is a refractory and fatal disease characterized by excessive pulmonary arterial cell remodeling. Uncontrolled proliferation and hypertrophy of pulmonary arterial smooth muscle cells (PASMCs), dysfunction of pulmonary arterial endothelial cells (PAECs), and abnormal perivascular infiltration of immune cells result in pulmonary arterial remodeling, followed by increased pulmonary vascular resistance and pulmonary pressure. Although various drugs targeting nitric oxide, endothelin-1 and prostacyclin pathways have been used in clinical settings, the mortality of pulmonary hypertension remains high. Multiple molecular abnormalities have been implicated in pulmonary hypertension, changes in numerous transcription factors have been identified as key regulators in pulmonary hypertension, and a role for pulmonary vascular remodeling has been highlighted. This review consolidates evidence linking transcription factors and their molecular mechanisms, from pulmonary vascular intima PAECs, vascular media PASMCs, and pulmonary arterial adventitia fibroblasts to pulmonary inflammatory cells. These findings will improve the understanding of particularly interactions between transcription factor-mediated cellular signaling pathways and identify novel therapies for pulmonary hypertension.

7.
Ann Transl Med ; 9(11): 926, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34350241

ABSTRACT

BACKGROUND: To explore the source, the role and the specific mechanism of IL-35 and its downstream molecules in the development of pulmonary hypertension. METHODS: 8-10 weeks male mice were undergoing hypoxia combined with SU5416 (HySu) to establish a pulmonary hypertension (PH) model. The phenotype of PH mice was measured by immunohistochemistry and immunofluorescence staining. The levels of two subunits (EBI3 and p35 subunits) in lung tissue were measured by real-time PCR and western blotting. EBI3 monoclonal antibody was administrated as IL-35 neutralization to offset systemic IL-35 expression. Fludarabine, an inhibitor of STAT1 (signal transducer and activator of transcription 1) was used to clarify the role of STAT1 under IL-35 treatment. RESULTS: After pulmonary hypertension, the expression of IL-35 and its two subunits (EBI3 and p35 subunits) in lung tissue were significantly increased. And the two subunits of IL-35 are highly expressed in Treg cells. Compared with the controlled PH mice, the IL-35 neutralization PH mice showed aggravated pulmonary hypertension phenotype. The specific manifestations are the increase of right ventricular systolic pressure (RVSP), the growing proportion of right heart [RV/(LV+S)], and the remodeling of pulmonary blood vessels increases. The expression of pulmonary vascular endothelium (CD31) in PH mice increased, and the proliferation ability of vascular endothelium enhanced after IL-35 was inhibited. IL-35 phosphorylates STAT1 through the receptor GP130 on pulmonary vascular endothelial cells, which in turn inhibits endothelial cell proliferation. IL-35 recombinant protein can reduce the expression of CD31 in lung tissues of PH mice. But the administration of STAT1 inhibitor made it invalid from the IL-35 effect of reversing pulmonary hypertension. CONCLUSIONS: Tregs-derived IL-35 can reverse the remodeling of pulmonary blood vessels and alleviate the progression of pulmonary hypertension by reducing the proliferation of endothelial cells.

8.
Cell Oncol (Dordr) ; 44(3): 643-659, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33646559

ABSTRACT

PURPOSE: Bone marrow stromal cells (BMSCs) have been implicated in multiple myeloma (MM) progression. However, the underlying mechanisms remain largely elusive. Therefore, we aimed to explore key factors in BMSCs that contribute to MM development. METHODS: RNA-sequencing was used to perform gene expression profiling in BMSCs. Enzyme-linked immunosorbent assays (ELISAs) were performed to determine the concentrations of PGE2 and TNFα in sera and conditioned media (CM). Western blotting, qRT-PCR and IHC were used to examine the expression of cyclooxygenase 2 (COX2) in BMSCs and to analyze the regulation of TNFα by COX2. Cell growth and adhesion assays were employed to explore the function of COX2 in vitro. A 5T33MMvt-KaLwRij mouse model was used to study the effects of COX2 inhibition in vivo. RESULTS: COX2 was found to be upregulated in MM patient-derived BMSCs and to play a critical role in BMSC-induced MM cell proliferation and adhesion. Administration of PGE2 to CM derived from BMSCs promoted MM cell proliferation and adhesion. Conversely, inhibition of COX2 in BMSCs greatly compromised BMSC-induced MM cell proliferation and adhesion. PCR array-based analysis of inflammatory cytokines indicated that COX2 upregulates the expression of TNFα. Subsequent rescue assays showed that an anti-TNFα monoclonal antibody could antagonize COX2-mediated MM cell proliferation and adhesion. Administration of NS398, a specific COX2 inhibitor, inhibited in vivo tumor growth and improved the survival of 5TMM mice. CONCLUSIONS: Our results indicate that COX2 contributes to BMSC-induced MM proliferation and adhesion by increasing the secretion of PGE2 and TNFα. Targeting COX2 in BMSCs may serve as a potential therapeutic approach of treating MM.


Subject(s)
Cyclooxygenase 2/metabolism , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/pathology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Adhesion/physiology , Cell Proliferation/physiology , Humans , Mice , Tumor Cells, Cultured
10.
J Pharmacol Exp Ther ; 373(2): 248-260, 2020 05.
Article in English | MEDLINE | ID: mdl-32165443

ABSTRACT

It has been identified that arginine vasopressin (AVP), vasopressin receptor 2(V2R), and the aquaporin 2 (AQP2) signaling pathway in the inner ear play important roles in hearing and balance functions through regulating the endolymph equilibrium; however, the contributions of this signaling pathway to the development of motion sickness are unclear. The present study was designed to investigate whether the activation of the AVP-V2R-AQP2 signaling pathway in the inner ear is involved in the induction of motion sickness and whether mozavaptan, a V2R antagonist, could reduce motion sickness. We found that both rotatory stimulus and intraperitoneal AVP injection induced conditioned taste aversion (a confirmed behavioral index for motion sickness) in rats and activated the AVP-V2R-AQP2 signaling pathway with a responsive V2R downregulation in the inner ears, and AVP perfusion in cultured epithelial cells from rat endolymphatic sacs induced similar changes in this pathway signaling. Vestibular training, V2R antagonist mozavaptan, or PKA inhibitor H89 blunted these changes in the V2R-AQP2 pathway signaling while reducing rotatory stimulus- or DDAVP (a V2R agonist)-induced motion sickness in rats and dogs. Therefore, our results suggest that activation of the inner ear AVP-V2R-AQP2 signaling pathway is potentially involved in the development of motion sickness; thus, mozavaptan targeting AVP V2Rs in the inner ear may provide us with a new application option to reduce motion sickness. SIGNIFICANCE STATEMENT: Motion sickness affects many people traveling or working. In the present study our results showed that activation of the inner ear arginine vasopressin-vaspopressin receptor 2 (V2R)-aquaporin 2 signaling pathway was potentially involved in the development of motion sickness and that blocking V2R with mozavaptan, a V2R antagonist, was much more effective in reducing motion sickness in both rat and dog; therefore, we demonstrated a new mechanism to underlie motion sickness and a new candidate drug to reduce motion sickness.


Subject(s)
Aquaporin 2/physiology , Arginine Vasopressin/physiology , Ear, Inner/physiology , Motion Sickness/etiology , Receptors, Vasopressin/physiology , Animals , Antidiuretic Hormone Receptor Antagonists/therapeutic use , Arginine Vasopressin/blood , Benzazepines/therapeutic use , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dogs , Female , Male , Motion Sickness/drug therapy , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
11.
Am J Respir Crit Care Med ; 201(10): 1263-1276, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31917615

ABSTRACT

Rationale: Vascular remodeling, including smooth muscle cell hypertrophy and proliferation, is the key pathological feature of pulmonary arterial hypertension (PAH). Prostaglandin I2 analogs (beraprost, iloprost, and treprostinil) are effective in the treatment of PAH. Of note, the clinically favorable effects of treprostinil in severe PAH may be attributable to concomitant activation of DP1 (D prostanoid receptor subtype 1).Objectives: To study the role of DP1 in the progression of PAH and its underlying mechanism.Methods: DP1 levels were examined in pulmonary arteries of patients and animals with PAH. Multiple genetic and pharmacologic approaches were used to investigate DP1-mediated signaling in PAH.Measurements and Main Results: DP1 expression was downregulated in hypoxia-treated pulmonary artery smooth muscle cells and in pulmonary arteries from rodent PAH models and patients with idiopathic PAH. DP1 deletion exacerbated pulmonary artery remodeling in hypoxia-induced PAH, whereas pharmacological activation or forced expression of the DP1 receptor had the opposite effect in different rodent models. DP1 deficiency promoted pulmonary artery smooth muscle cell hypertrophy and proliferation in response to hypoxia via induction of mTORC1 (mammalian target of rapamycin complex 1) activity. Rapamycin, an inhibitor of mTORC1, alleviated the hypoxia-induced exacerbation of PAH in DP1-knockout mice. DP1 activation facilitated raptor dissociation from mTORC1 and suppressed mTORC1 activity through PKA (protein kinase A)-dependent phosphorylation of raptor at Ser791. Moreover, treprostinil treatment blocked the progression of hypoxia-induced PAH in mice in part by targeting the DP1 receptor.Conclusions: DP1 activation attenuates hypoxia-induced pulmonary artery remodeling and PAH through PKA-mediated dissociation of raptor from mTORC1. These results suggest that the DP1 receptor may serve as a therapeutic target for the management of PAH.


Subject(s)
Hypoxia/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Pulmonary Arterial Hypertension/genetics , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Vascular Remodeling/genetics , Animals , Antihypertensive Agents/pharmacology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Humans , Hypertrophy , Immunosuppressive Agents/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery , RNA, Messenger/metabolism , Rats , Sirolimus/pharmacology
12.
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
13.
Mol Carcinog ; 59(3): 265-280, 2020 03.
Article in English | MEDLINE | ID: mdl-31867776

ABSTRACT

Nasopharyngeal carcinoma (NPC) has the highest rate of metastasis among head and neck cancers, and distant metastasis is the major reason for treatment failure. We have previously shown that high cyclooxygenase-2 (COX-2) expression is associated with a poor prognosis of patients with NPC and inhibits chemotherapy-induced senescence in NPC cells. In this study, we found that COX-2 was upregulated in cancer-associated fibroblasts (CAFs) derived from NPC by RNA-Seq. Furthermore, elevated COX-2 expression in CAF was detected in NPC patients with poor survival and distant metastasis by using immunohistochemistry. Then, we identified that COX-2 is highly expressed in CAF at the distant metastasis site in seven paired NPC patients. High expression of COX-2 and secretion of prostaglandin E2, a major product catalyzed by COX-2 in fibroblasts, promotes migration and invasiveness of NPC cells in vitro. On the contrary, inhibition of COX-2 has the opposite effect in vitro as well as in the COX-2-/- mouse with the lung metastasis model in vivo. Mechanistically, we discovered that COX-2 elevates tumor necrosis factor-α expression in CAF to promote NPC cell migration and invasiveness. Overall, our results identified a novel target in CAF promoting NPC metastasis. Our findings suggested that high expression of COX-2 in CAF may serve as a new prognostic indicator for NPC metastasis and provide the possibility of targeting CAF for treating advanced NPC.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Cyclooxygenase 2/genetics , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Movement , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Nasopharyngeal Carcinoma/diagnosis , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prognosis , Up-Regulation
14.
J Pharmacol Exp Ther ; 370(3): 416-426, 2019 09.
Article in English | MEDLINE | ID: mdl-31248980

ABSTRACT

Inflammatory bowel disease (IBD) is associated with an increased risk for thromboembolism, platelet activation, and abnormalities in platelet number and size. In colitis, platelets can extravasate into the colonic interstitium. We generated a mouse with a specific deletion of cyclooxygenase (COX)-1 in megakaryocytes/platelets [(COX-1 conditional knockout (cKO)] to clarify the role of platelet activation in the development of inflammation and fibrosis in dextran sodium sulfate (DSS)-induced colitis. The disease activity index was assessed, and colonic specimens were evaluated for histologic features of epithelial barrier damage, inflammation, and fibrosis. Cocultures of platelets and myofibroblasts were performed. We found that the specific deletion of COX-1 in platelets, which recapitulated the human pharmacodynamics of low-dose aspirin, that is, suppression of platelet thromboxane (TX)A2 production associated with substantial sparing of the systemic production of prostacyclin, resulted in milder symptoms of colitis, in the acute phase, and almost complete recovery from the disease after DSS withdrawal. Reduced colonic accumulation of macrophages and myofibroblasts and collagen deposition was found. Platelet-derived TXA2 enhanced the ability of myofibroblasts to proliferate and migrate in vitro, and these effects were prevented by platelet COX-1 inhibition or antagonism of the TXA2 receptor. Our findings allow a significant advance in the knowledge of the role of platelet-derived TXA2 in the development of colitis and fibrosis in response to intestinal damage and provide the rationale to investigate the potential efficacy of the antiplatelet agent low-dose aspirin in limiting the inflammatory response and fibrosis associated with IBD. SIGNIFICANCE STATEMENT: Inflammatory bowel disease (IBD) is characterized by the development of a chronic inflammatory response, which can lead to intestinal fibrosis for which currently there is no medical treatment. Through the generation of a mouse with specific deletion of cyclooxygenase-1 in megakaryocytes/platelets, which recapitulates the human pharmacodynamics of low-dose aspirin, we demonstrate the important role of platelet-derived thromboxane A2 in the development of experimental colitis and fibrosis, thus providing the rationale to investigate the potential efficacy of low-dose aspirin in limiting the inflammation and tissue damage associated with IBD.


Subject(s)
Blood Platelets/metabolism , Colitis/chemically induced , Colitis/enzymology , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Dextran Sulfate/pharmacology , Gene Deletion , Animals , Blood Platelets/drug effects , Blood Platelets/pathology , Colitis/blood , Colitis/genetics , Colon/drug effects , Colon/metabolism , Colon/pathology , Humans , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Myofibroblasts/drug effects , Myofibroblasts/pathology , Prostaglandins/biosynthesis
15.
FASEB J ; 33(9): 10207-10217, 2019 09.
Article in English | MEDLINE | ID: mdl-31216422

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is an environmental pollutant that causes cardiovascular toxicity. The phenotypic transformation of vascular smooth muscle cells (VSMCs) from the contractile to the synthetic phenotype is a hallmark of vascular response to injury. However, the precise role and molecular mechanism of TCDD in vascular remodeling remains unknown. In the present study, we found that TCDD treatment promoted VSMC phenotypic transition from contractile to synthetic phenotype and exaggerated vascular neointimal hyperplasia after wire injury in mice. TCDD treatment enhanced VSMC entry into cell cycle from G0/G1 phase to S and G2/M phase. The expression of cyclin D1, cyclin-dependent kinase 4 (CDK4), and its phosphorylation were coordinately increased in response to TCDD treatment. Knocking down of aryl hydrocarbon receptor (AHR) inhibited VSMC phenotypic transition induced by TCDD and promoted S/G2 phase cell cycle arrest. TCDD treatment markedly increased oncogenic c-Jun gene expression in VSMCs. ChIP assay revealed the direct binding of AHR on the promoter of c-Jun to up-regulate the mRNA expression of c-Jun. Silencing of c-Jun gene enhanced the expression of p53 and p21, whereas attenuated the expression of CDK4 and cyclin D1 leading to the decrease in the TCDD-stimulated VSMC proliferation and synthetic phenotype transition in vitro. In vivo study showed that genetic ablation of c-Jun in VSMCs restricted injury-induced neointimal hyperplasia in TCDD-treated mice. Thus, TCDD exposure exaggerated injury-induced vascular remodeling by the activation of AHR and up-regulation of the expression of its target gene c-Jun, indicating that inhibition of AHR may be a promising prevention strategy for TCDD-associated cardiovascular diseases.-Guo, S., Zhang, R., Liu, Q., Wan, Q., Wang, Y., Yu, Y., Liu, G., Shen, Y., Yu, Y., Zhang, J. 2,3,7,8-Tetrachlorodibenzo-p-dioxin promotes injury-induced vascular neointima formation in mice.


Subject(s)
Endothelium, Vascular/injuries , Environmental Pollutants/toxicity , Neointima/chemically induced , Polychlorinated Dibenzodioxins/toxicity , Animals , Aorta/cytology , Cell Cycle/drug effects , Endothelium, Vascular/drug effects , Femoral Artery/drug effects , Femoral Artery/injuries , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Genes, Reporter , Genes, jun , Humans , Hyperplasia , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neointima/etiology , Phenotype , Proto-Oncogene Proteins c-jun/biosynthesis , Proto-Oncogene Proteins c-jun/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/physiology , Vascular Remodeling/drug effects
16.
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
17.
Int J Oncol ; 53(3): 1138-1148, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29956730

ABSTRACT

Resistance to radiotherapy and chemotherapy currently represents one of the major reasons for therapeutic failure in nasopharyngeal carcinoma (NPC). However, the mechanisms underlying resistance to chemotherapy in NPC remain unclear. In this study, cell counting assay, cell cycle assay and senescence associated ß-galactosidase activity were performed to evaluate cell growth, proliferation and senescence, respectively. We found that the aberrant expression of cyclooxygenase-2 (COX-2) was associated with a poor outcome and recurrance in patients with NPC. In NPC cells, COX-2 overexpression increased cell proliferation, inhibited cellular senescence and resulted in chemoresistance, while the knockdown of COX-2 reduced cell proliferation, promoted cellular senescence and overcame chemoresistance. Furthermore, fibroblasts from COX-2 knockout mice exhibited cellular senescence, particularly when treated with chemotherapeutic agents. Mechanistically, COX-2 interacted with p53 protein and inhibited cellular senescence, which resulted in chemotherapeutic resistance. On the whole, these findings indicate that COX-2 may play a critical role in chemotherapeutic resistance in NPC via the inhibition of chemotherapy-induced senescence via the inactivation of p53. This study provides experimental evidence for the preclinical value of increasing chemotherapy-induced senescence by targeting COX-2 as an effective antitumor treatment in patients with recurrent NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Cellular Senescence/drug effects , Cyclooxygenase 2/metabolism , Nasopharyngeal Neoplasms/drug therapy , Neoplasm Recurrence, Local/pathology , Animals , Antineoplastic Agents/therapeutic use , Benzothiazoles/pharmacology , Biomarkers, Tumor , Carcinoma/mortality , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Cyclooxygenase 2/genetics , Drug Resistance, Neoplasm , Female , Fibroblasts , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/pathology , Neoplasm Recurrence, Local/epidemiology , Primary Cell Culture , RNA Interference , RNA, Small Interfering/metabolism , Specific Pathogen-Free Organisms , Survival Analysis , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
18.
FASEB J ; 32(10): 5413-5425, 2018 10.
Article in English | MEDLINE | ID: mdl-29723062

ABSTRACT

Mechanical insults, such as stent implantation, can induce endothelial injury, vascular inflammation, and ultimately lead to vascular neointimal hyperplasia. Resolvin E1 (RvE1), derived from the ω3 fatty acid eicosapentaenoic acid, can facilitate the resolution of inflammation in many settings. We therefore aimed to determine if there was a role for RvE1 in preventing neointimal formation after arterial injury and to understand the underlying mechanisms. Vascular inflammation and neointimal hyperplasia were induced by wire injury in the femoral arteries of mice. Administration of exogenous RvE1 and endogenously generated RvE1 via dietary supplementation with eicosapentaenoic acid and aspirin markedly reduced vascular neointima formation in this model. Mechanistically, RvE1 was found to inhibit vascular neutrophil infiltration, promote macrophage polarization toward an M2-like phenotype, suppress T-cell trafficking by reducing RANTES secretion from vascular smooth muscle cells, and inhibit vascular smooth muscle cell migration. In summary, RvE1 demonstrated a protective role against vascular inflammation and remodeling in response to mechanical injury, suggesting that it may serve as an adjuvant therapeutic agent for percutaneous coronary interventions, such as stent implantation.-Liu, G., Gong, Y., Zhang, R., Piao, L., Li, X., Liu, Q., Yan, S., Shen, Y., Guo, S., Zhu, M., Yin, H., Funk, C. D., Zhang, J., Yu, Y. Resolvin E1 attenuates injury-induced vascular neointimal formation by inhibition of inflammatory responses and vascular smooth muscle cell migration.


Subject(s)
Cell Movement/drug effects , Eicosapentaenoic Acid/analogs & derivatives , Femoral Artery , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/prevention & control , Animals , Disease Models, Animal , Eicosapentaenoic Acid/pharmacology , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neointima/metabolism , Neointima/pathology
19.
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
20.
J Cell Mol Med ; 21(12): 3540-3551, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28699682

ABSTRACT

Endothelial injury or dysfunction is an early event in the pathogenesis of atherosclerosis. Epidemiological and animal studies have shown that 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) exposure increases morbidity and mortality from chronic cardiovascular diseases, including atherosclerosis. However, whether or how TCDD exposure causes endothelial injury or dysfunction remains largely unknown. Cultured human umbilical vein endothelial cells (HUVECs) were exposed to different doses of TCDD, and cell apoptosis was examined. We found that TCDD treatment increased caspase 3 activity and apoptosis in HUVECs in a dose-dependent manner,at doses from 10 to 40 nM. TCDD increased cyclooxygenase enzymes (COX)-2 expression and its downstream prostaglandin (PG) production (mainly PGE2 and 6-keto-PGF1α ) in HUVECs. Interestingly, inhibition of COX-2, but not COX-1, markedly attenuated TCDD-triggered apoptosis in HUVECs. Pharmacological inhibition or gene silencing of the PGE2 receptor subtype 3 (EP3) suppressed the augmented apoptosis in TCDD-treated HUVECs. Activation of the EP3 receptor enhanced p38 MAPK phosphorylation and decreased Bcl-2 expression following TCDD treatment. Both p38 MAPK suppression and Bcl-2 overexpression attenuated the apoptosis in TCDD-treated HUVECs. TCDD increased EP3-dependent Rho activity and subsequently promoted p38MAPK/Bcl-2 pathway-mediated apoptosis in HUVECs. In addition, TCDD promoted apoptosis in vascular endothelium and delayed re-endothelialization after femoral artery injury in wild-type (WT) mice, but not in EP3-/- mice. In summary, TCDD promotes endothelial apoptosis through the COX-2/PGE2 /EP3/p38MAPK/Bcl-2 pathway. Given the cardiovascular hazard of a COX-2 inhibitor, our findings indicate that the EP3 receptor and its downstream pathways may be potential targets for prevention of TCDD-associated cardiovascular diseases.


Subject(s)
Endothelium, Vascular/drug effects , Environmental Pollutants/toxicity , Polychlorinated Dibenzodioxins/toxicity , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Prostaglandin E, EP3 Subtype/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/agonists , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Femoral Artery/injuries , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Re-Epithelialization/physiology , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...