Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Commun Biol ; 5(1): 1192, 2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36344664

RESUMO

Pulmonary arterial hypertension (PAH) is an unmet clinical need. The lack of models of human disease is a key obstacle to drug development. We present a biomimetic model of pulmonary arterial endothelial-smooth muscle cell interactions in PAH, combining natural and induced bone morphogenetic protein receptor 2 (BMPR2) dysfunction with hypoxia to induce smooth muscle activation and proliferation, which is responsive to drug treatment. BMPR2- and oxygenation-specific changes in endothelial and smooth muscle gene expression, consistent with observations made in genomic and biochemical studies of PAH, enable insights into underlying disease pathways and mechanisms of drug response. The model captures key changes in the pulmonary endothelial phenotype that are essential for the induction of SMC remodelling, including a BMPR2-SOX17-prostacyclin signalling axis and offers an easily accessible approach for researchers to study pulmonary vascular remodelling and advance drug development in PAH.


Assuntos
Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Fatores de Transcrição SOXF , Humanos , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Epoprostenol/genética , Epoprostenol/metabolismo , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Hipertensão Arterial Pulmonar/genética , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo
2.
J Clin Invest ; 131(7)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33529171

RESUMO

Tregs restrain both the innate and adaptive immune systems to maintain homeostasis. Allergic airway inflammation, characterized by a Th2 response that results from a breakdown of tolerance to innocuous environmental antigens, is negatively regulated by Tregs. We previously reported that prostaglandin I2 (PGI2) promoted immune tolerance in models of allergic inflammation; however, the effect of PGI2 on Treg function was not investigated. Tregs from mice deficient in the PGI2 receptor IP (IP KO) had impaired suppressive capabilities during allergic airway inflammatory responses compared with mice in which PGI2 signaling was intact. IP KO Tregs had significantly enhanced expression of immunoglobulin-like transcript 3 (ILT3) compared with WT Tregs, which may contribute to the impairment of the IP KO Treg's ability to suppress Th2 responses. Using fate-mapping mice, we reported that PGI2 signaling prevents Treg reprogramming toward a pathogenic phenotype. PGI2 analogs promoted the differentiation of naive T cells to Tregs in both mice and humans via repression of ß-catenin signaling. Finally, a missense variant in IP in humans was strongly associated with chronic obstructive asthma. Together, these data support that PGI2 signaling licenses Treg suppressive function and that PGI2 is a therapeutic target for enhancing Treg function.


Assuntos
Asma/imunologia , Reprogramação Celular/imunologia , Epoprostenol/imunologia , Tolerância Imunológica , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Asma/genética , Asma/patologia , Reprogramação Celular/genética , Doença Crônica , Epoprostenol/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/imunologia , Transdução de Sinais/genética , Linfócitos T Reguladores/patologia
3.
EMBO J ; 39(14): e103454, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32484988

RESUMO

The alarm cytokine interleukin-1ß (IL-1ß) is a potent activator of the inflammatory cascade following pathogen recognition. IL-1ß production typically requires two signals: first, priming by recognition of pathogen-associated molecular patterns leads to the production of immature pro-IL-1ß; subsequently, inflammasome activation by a secondary signal allows cleavage and maturation of IL-1ß from its pro-form. However, despite the important role of IL-1ß in controlling local and systemic inflammation, its overall regulation is still not fully understood. Here we demonstrate that peritoneal tissue-resident macrophages use an active inhibitory pathway, to suppress IL-1ß processing, which can otherwise occur in the absence of a second signal. Programming by the transcription factor Gata6 controls the expression of prostacyclin synthase, which is required for prostacyclin production after lipopolysaccharide stimulation and optimal induction of IL-10. In the absence of secondary signal, IL-10 potently inhibits IL-1ß processing, providing a previously unrecognized control of IL-1ß in tissue-resident macrophages.


Assuntos
Epoprostenol/imunologia , Interleucina-10/imunologia , Interleucina-1beta/imunologia , Macrófagos Peritoneais/imunologia , Animais , Epoprostenol/genética , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-10/genética , Interleucina-1beta/genética , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Transgênicos
4.
J Neuroimmune Pharmacol ; 15(2): 292-308, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31897976

RESUMO

Cellular arachidonic acid (AA), an unsaturated fatty acid found ubiquitously in plasma membranes, is metabolized to different prostanoids, such as prostacyclin (PGI2) and prostaglandin E2 (PGE2), by the three-step reactions coupling the upstream cyclooxygenase (COX) isoforms (COX-1 and COX-2) with the corresponding individual downstream synthases. While the vascular actions of these prostanoids are well-characterized, their specific roles in the hippocampus, a major brain area for memory, are poorly understood. The major obstacle for its understanding in the brain was to mimic the biosynthesis of each prostanoid. To solve the problem, we utilized Single-Chain Hybrid Enzyme Complexes (SCHECs), which could successfully control cellular AA metabolites to the desired PGI2 or PGE2. Our in vitro studies suggested that neurons with higher PGI2 content and lower PGE2 content exhibited survival protection and resistance to Amyloid-ß-induced neurotoxicity. Further extending to an in vivo model, the hybrid of PGI2-producing transgenic mice and Alzheimer's disease (AD) mice showed restored long-term memory. These findings suggested that the vascular prostanoids, PGI2 and PGE2, exerted significant regulatory influences on neuronal protection (by PGI2), or damage (by PGE2) in the hippocampus, and raised a concern that the wide uses of aspirin in cardiovascular diseases may exert negative impacts on neurodegenerative protection. Graphic Abstract Our study intended to understand the crosstalk of prostanoids in the hippocampus, a major brain area impacted in AD, by using hybrid enzymes to redirect the synthesis of prostanoids to PGE2 and PGI2, respectively. Our data indicated that during inflammation, the vascular mediators, PGI2 and PGE2, exerted significant regulatory influences on neuronal protection (by PGI2), or damage (by PGE2) in the hippocampus. These findings also raised a concern that the widely uses of non-steroidal anti-inflammatory drugs in cardiovascular diseases may exert negative impacts on neurodegenerative protection.


Assuntos
Epoprostenol/biossíntese , Hipocampo/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Regulação para Cima/fisiologia , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Epoprostenol/genética , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Iloprosta/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Regulação para Cima/efeitos dos fármacos
5.
Circ Res ; 125(9): 847-854, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31510878

RESUMO

RATIONALE: Endothelial cells (ECs) and platelets, which respectively produce antithrombotic prostacyclin and prothrombotic thromboxane A2, both express COX1 (cyclooxygenase1). Consequently, there has been no way to delineate any antithrombotic role for COX1-derived prostacyclin from the prothrombotic effects of platelet COX1. By contrast, an antithrombotic role for COX2, which is absent in platelets, is straightforward to demonstrate. This has resulted in an incomplete understanding of the relative importance of COX1 versus COX2 in prostacyclin production and antithrombotic protection in vivo. OBJECTIVE: We sought to identify the role, if any, of COX1-derived prostacyclin in antithrombotic protection in vivo and compare this to the established protective role of COX2. METHODS AND RESULTS: We developed vascular-specific COX1 knockout mice and studied them alongside endothelial-specific COX2 knockout mice. COX1 immunoreactivity and prostacyclin production were primarily associated with the endothelial layer of aortae; freshly isolated aortic ECs released >10-fold more prostacyclin than smooth muscle cells. Moreover, aortic prostacyclin production, the ability of aortic rings to inhibit platelet aggregation and plasma prostacyclin levels were reduced when COX1 was knocked out in ECs but not in smooth muscle cells. When thrombosis was measured in vivo after FeCl3 carotid artery injury, endothelial COX1 deletion accelerated thrombosis to a similar extent as prostacyclin receptor blockade. However, this effect was lost when COX1 was deleted from both ECs and platelets. Deletion of COX2 from ECs also resulted in a prothrombotic phenotype that was independent of local vascular prostacyclin production. CONCLUSIONS: These data demonstrate for the first time that, in healthy animals, endothelial COX1 provides an essential antithrombotic tone, which is masked when COX1 activity is lost in both ECs and platelets. These results help us define a new 2-component paradigm wherein thrombotic tone is regulated by both COX1 and COX2 through complementary but mechanistically distinct pathways.


Assuntos
Ciclo-Oxigenase 1/deficiência , Endotélio Vascular/metabolismo , Epoprostenol/metabolismo , Fibrinolíticos/metabolismo , Deleção de Genes , Proteínas de Membrana/deficiência , Agregação Plaquetária/fisiologia , Animais , Aorta/metabolismo , Ciclo-Oxigenase 1/genética , Epoprostenol/genética , Feminino , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos
6.
J Cell Physiol ; 233(10): 6386-6394, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29741779

RESUMO

Atherosclerosis is identified as the formation of atherosclerotic plaques, which could initiate the formation of a blood clot in which its growth to coronary artery can lead to a heart attack. N-methyltransferase (NNMT) is an enzyme that converts the NAM (nicotinamide) to its methylated form, N1-methylnicotinamide (MNAM). Higher levels of MNAM have been reported in cases with coronary artery disease (CAD). Further, MNAM increases endothelial prostacyclin (PGI2) and nitric oxide (NO) and thereby causes vasorelaxation. The vasoprotective, anti-inflammatory and anti-thrombotic roles of MNAM have been well documented; however, the exact underlying mechanisms remain to be clarified. Due to potential role of MNAM in the formation of lipid droplets (LDs), it might exert its function in coordination with lipids, and their targets. In this study, we summarized the roles of MNAM in cardiovascular system and highlighted its possible mode of actions.


Assuntos
Aterosclerose/genética , Sistema Cardiovascular/metabolismo , Doença da Artéria Coronariana/genética , Niacinamida/análogos & derivados , Aterosclerose/metabolismo , Doença da Artéria Coronariana/metabolismo , Epoprostenol/genética , Epoprostenol/metabolismo , Humanos , Gotículas Lipídicas/metabolismo , Niacinamida/genética , Niacinamida/metabolismo , Óxido Nítrico/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-29660395

RESUMO

Endogenous prostaglandin I2 (PGI2) has inhibitory effects on immune responses against pathogens or allergens; however, the immunomodulatory activity of endogenous PGI2 signaling in endotoxin-induced inflammation is unknown. To test the hypothesis that endogenous PGI2 down-regulates endotoxin-induced lung inflammation, C57BL/6 wild type (WT) and PGI2 receptor (IP) KO mice were challenged intranasally with LPS. Urine 6-keto-PGF1α, a stable metabolite of PGI2, was significantly increased following the LPS-challenge, suggesting that endogenous PGI2 signaling modulates the host response to LPS-challenge. IPKO mice had a significant increase in neutrophils in the BAL fluid as well as increased proteins of KC, LIX, and TNF-α in lung homogenates compared with WT mice. In contrast, IL-10 was decreased in LPS-challenged IPKO mice compared with WT mice. The PGI2 analog cicaprost significantly decreased LPS-induced KC, and TNF-α, but increased IL-10 and AREG in bone marrow-derived dendritic cells (BMDCs) and bone marrow-derived macrophages (BMMs) compared with vehicle-treatment. These results indicated that endogenous PGI2 signaling attenuated neutrophilic lung inflammation through the reduced inflammatory cytokine and chemokine and enhanced IL-10.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Epoprostenol/metabolismo , Lipopolissacarídeos/toxicidade , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Transdução de Sinais , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/patologia , Animais , Modelos Animais de Doenças , Epoprostenol/genética , Camundongos , Camundongos Knockout , Neutrófilos/patologia
8.
Hum Gene Ther ; 28(8): 681-689, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28530128

RESUMO

Pulmonary arterial hypertension (PAH) is a progressive disease that culminates in right heart failure and death. Prostacyclin (PGI2) and its derivatives are effective treatments for PAH when administered as continuous parenteral infusions. This treatment paradigm requires medical sophistication, and patients are at risk for complications from an indewelling catheter; drug interruptions may result in rebound pulmonary hypertension and death. We hypothesized that the salivary gland can be repurposed into an endogenous production site for circulating PGI2 through the expression of a fusion protein embodying cyclooxygenase-1 (Cox1) and prostacyclin synthase (PGIS) domains. We utilized ultrasound-assisted gene transfer, a nonviral gene transfer strategy that achieves robust gene transfer to the salivary gland. We initially found that Cox1-PGIS expression in livers of mice using an adenoviral vector dramatically increased circulating PGI2 relative to untreated rats or rats treated with PGIS alone. We then utilized ultrasound-assisted gene transfer to express Cox1-PGIS in the submandibular glands of rats and showed a significant elevation of circulating PGI2 that corresponded to approximately 30% of that seen in humans undergoing intravenous infusion therapy for PAH. These results suggest the feasibility of gene therapy to drive endogenous biosynthesis of PGI2 as a therapeutic strategy for the treatment of PAH.


Assuntos
Ciclo-Oxigenase 1/genética , Epoprostenol/genética , Expressão Gênica , Técnicas de Transferência de Genes , Proteínas Recombinantes de Fusão/genética , Glândulas Salivares/metabolismo , Adenoviridae/genética , Animais , DNA Complementar/genética , DNA Complementar/metabolismo , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Fígado/metabolismo , Masculino , Camundongos , Ratos , Proteínas Recombinantes de Fusão/sangue , Proteínas Recombinantes de Fusão/metabolismo , Glândula Submandibular/metabolismo , Fatores de Tempo , Transcrição Gênica
9.
J Clin Invest ; 125(9): 3606-18, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26280574

RESUMO

Mesenchymal stem cell (MSC) therapy is an emerging field of regenerative medicine; however, it is often unclear how these cells mediate repair. Here, we investigated the use of MSCs in the treatment of intestinal disease and modeled abnormal repair by creating focal wounds in the colonic mucosa of prostaglandin-deficient mice. These wounds developed into ulcers that infiltrated the outer intestinal wall. We determined that penetrating ulcer formation in this model resulted from increased hypoxia and smooth muscle wall necrosis. Prostaglandin I2 (PGI2) stimulated VEGF-dependent angiogenesis to prevent penetrating ulcers. Treatment of mucosally injured WT mice with a VEGFR inhibitor resulted in the development of penetrating ulcers, further demonstrating that VEGF is critical for mucosal repair. We next used this model to address the role of transplanted colonic MSCs (cMSCs) in intestinal repair. Compared with intravenously injected cMSCs, mucosally injected cMSCs more effectively prevented the development of penetrating ulcers, as they were more efficiently recruited to colonic wounds. Importantly, mucosally injected cMSCs stimulated angiogenesis in a VEGF-dependent manner. Together, our results reveal that penetrating ulcer formation results from a reduction of local angiogenesis and targeted injection of MSCs can optimize transplantation therapy. Moreover, local MSC injection has potential for treating diseases with features of abnormal angiogenesis and repair.


Assuntos
Colo , Mucosa Intestinal , Transplante de Células-Tronco Mesenquimais , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , Ferimentos e Lesões , Aloenxertos , Animais , Colo/lesões , Colo/metabolismo , Colo/patologia , Epoprostenol/genética , Epoprostenol/metabolismo , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Fator A de Crescimento do Endotélio Vascular/genética , Ferimentos e Lesões/genética , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia , Ferimentos e Lesões/terapia
10.
FASEB J ; 29(11): 4568-78, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26183771

RESUMO

Eicosanoids are important vascular regulators, but the phospholipase A2 (PLA2) isoforms supporting their production within the cardiovascular system are not fully understood. To address this, we have studied platelets, endothelial cells, and leukocytes from 2 siblings with a homozygous loss-of-function mutation in group IVA cytosolic phospholipase A2 (cPLA2α). Chromatography/mass spectrometry was used to determine levels of a broad range of eicosanoids produced by isolated vascular cells, and in plasma and urine. Eicosanoid release data were paired with studies of cellular function. Absence of cPLA2α almost abolished eicosanoid synthesis in platelets (e.g., thromboxane A2, control 20.5 ± 1.4 ng/ml vs. patient 0.1 ng/ml) and leukocytes [e.g., prostaglandin E2 (PGE2), control 21.9 ± 7.4 ng/ml vs. patient 1.9 ng/ml], and this was associated with impaired platelet activation and enhanced inflammatory responses. cPLA2α-deficient endothelial cells showed reduced, but not absent, formation of prostaglandin I2 (prostacyclin; control 956 ± 422 pg/ml vs. patient 196 pg/ml) and were primed for inflammation. In the urine, prostaglandin metabolites were selectively influenced by cPLA2α deficiency. For example, prostacyclin metabolites were strongly reduced (18.4% of control) in patients lacking cPLA2α, whereas PGE2 metabolites (77.8% of control) were similar to healthy volunteer levels. These studies constitute a definitive account, demonstrating the fundamental role of cPLA2α to eicosanoid formation and cellular responses within the human circulation.


Assuntos
Antígenos de Plaquetas Humanas/genética , Plaquetas/enzimologia , Dinoprostona/genética , Células Endoteliais/enzimologia , Epoprostenol/genética , Leucócitos/enzimologia , Mutação , Adulto , Plaquetas/patologia , Dinoprostona/biossíntese , Células Endoteliais/patologia , Epoprostenol/biossíntese , Feminino , Humanos , Leucócitos/patologia , Masculino , Ativação Plaquetária/genética
11.
Blood ; 125(4): 710-9, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25339356

RESUMO

The precise mechanism for reduced thrombosis in prekallikrein null mice (Klkb1(-/-)) is unknown. Klkb1(-/-) mice have delayed carotid artery occlusion times on the rose bengal and ferric chloride thrombosis models. Klkb1(-/-) plasmas have long-activated partial thromboplastin times and defective contact activation-induced thrombin generation that partially corrects upon prolonged incubation. However, in contact activation-induced pulmonary thromboembolism by collagen/epinephrine or long-chain polyphosphate, Klkb1(-/-) mice, unlike F12(-/-) mice, do not have survival advantage. Klkb1(-/-) mice have reduced plasma BK levels and renal B2R mRNA. They also have increased expression of the renal receptor Mas and plasma prostacyclin. Increased prostacyclin is associated with elevated aortic vasculoprotective transcription factors Sirt1 and KLF4. Treatment of Klkb1(-/-) mice with the Mas antagonist A-779, COX-2 inhibitor nimesulide, or Sirt1 inhibitor splitomicin lowers plasma prostacyclin and normalizes arterial thrombosis times. Treatment of normal mice with the Mas agonist AVE0991 reduces thrombosis. Klkb1(-/-) mice have reduced aortic tissue factor (TF) mRNA, antigen, and activity. In sum, Klkb1(-/-) mice have a novel mechanism for thrombosis protection in addition to reduced contact activation. This pathway arises when bradykinin delivery to vasculature is compromised and mediated by increased receptor Mas, prostacyclin, Sirt1, and KLF4, leading to reduced vascular TF.


Assuntos
Trombose das Artérias Carótidas , Epoprostenol , Fatores de Transcrição Kruppel-Like , Pré-Calicreína , Proteínas Proto-Oncogênicas , Receptores Acoplados a Proteínas G , Tromboplastina , Angiotensina II/análogos & derivados , Angiotensina II/farmacologia , Animais , Trombose das Artérias Carótidas/induzido quimicamente , Trombose das Artérias Carótidas/genética , Trombose das Artérias Carótidas/metabolismo , Trombose das Artérias Carótidas/patologia , Epoprostenol/biossíntese , Epoprostenol/genética , Imidazóis/farmacologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Naftalenos/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Tempo de Tromboplastina Parcial , Fragmentos de Peptídeos/farmacologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Pironas/farmacologia , RNA Mensageiro , Receptor B2 da Bradicinina/biossíntese , Receptor B2 da Bradicinina/genética , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/biossíntese , Sirtuína 1/genética , Sulfonamidas/farmacologia , Sinaptotagminas/biossíntese , Sinaptotagminas/genética , Tromboplastina/antagonistas & inibidores , Tromboplastina/biossíntese , Tromboplastina/genética
12.
Int J Mol Sci ; 15(2): 2826-39, 2014 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-24557578

RESUMO

The selection of vascular grafts for coronary artery bypass surgery is crucial for a positive outcome. This study aimed to establish a novel line of vascular endothelial cells with a potent anticoagulant effect. A lentiviral vector was used to stably transfect human umbilical vein endothelial cells (HUVECs) with PGI2S alone (HUVEC-PGI2S) or both PGI2S and tPA (HUVEC-PGI2S-tPA). Both HUVEC-PGI2S and HUVEC-PGI2S-tPA cells over-expressing PGI2S and tPA were compared to mock-transfected cells. The enzyme-linked immuno sorbent assay (ELISAs) demonstrated that the anticoagulation components, ATIII and PLG, were up-regulated and coagulation factor FVIII was down-regulated in both cell lines. QRT-PCR and western blotting demonstrated the vasodilation and platelet disaggregation proteins PKA, PKC, and PTGIR were up-regulated in both cell lines, but MAPK expression was not altered in either cell line. However, cell viability and colony formation assays and cell cycle analysis demonstrated that both cell lines had a lower rate of cell growth and induced G1 phase arrest. HUVEC-PGI2S and HUVEC-PGI2S-tPA cells have a potent anticoagulant effect and their use in vascular heterografts may decrease the risk of thrombosis.


Assuntos
Anticoagulantes/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Epoprostenol/metabolismo , Proteína Quinase C/metabolismo , Ativador de Plasminogênio Tecidual/metabolismo , Antitrombina III/metabolismo , Sobrevivência Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Regulação para Baixo , Epoprostenol/genética , Fator VIII/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Agregação Plaquetária/efeitos dos fármacos , Proteína Quinase C/genética , Receptores de Epoprostenol , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ativador de Plasminogênio Tecidual/genética , Regulação para Cima
13.
J Mol Endocrinol ; 52(2): 215-22, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24444496

RESUMO

Recent studies have reported that subclinical hypothyroidism (SCH) is associated with atherosclerosis (AS). Thyroid hormone is maintained at normal levels in patients with SCH, whereas TSH is increased. However, the pathogenesis of AS in association with SCH is only partially understood. In addition, endothelial dysfunction plays an important role in the development of AS. The purpose of the present research was to study the direct effect of TSH on human umbilical vein endothelial cells (HUVECs). The expression of some genes associated with endothelial dysfunction after treatment with TSH was evaluated by real-time PCR and western blotting respectively. At first, we showed that the TSH receptor (TSHR) is expressed in HUVECs. We also provide evidence indicating that TSH treatment promotes tumor necrosis factor α-induced endothelial cells interactions by upregulating the expression of the adhesion molecules intercellular adhesion molecule-1. Furthermore, the expression of endothelial nitric oxide synthase (eNOS) and prostacyclin (PGI2) was significantly attenuated following treatment with TSH in dose- and time-dependent manner. Conversely, the results indicated that TSH upregulated endothelin-1 (ET1) mRNA and protein expression in HUVECs, similar effects were observed for plasminogen activator inhibitor-1 (PAI1) after treatment with various concentrations of TSH. Taken together, these results demonstrate that elevated TSH can promote endothelial dysfunction by altering gene expression in HUVECs.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Tireotropina/farmacologia , Animais , Bovinos , Regulação para Baixo/efeitos dos fármacos , Endotelina-1/genética , Endotelina-1/metabolismo , Epoprostenol/genética , Epoprostenol/metabolismo , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Tireotropina/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/efeitos dos fármacos
14.
J Ethnopharmacol ; 151(1): 176-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24269248

RESUMO

AIM OF THE STUDY: Toona sinensis Roem. (Meliaceae; Toona sinensis; Chinese toon) is a type of arbor that is widely distributed in Asia. The fruits of Toona sinensis Roem has been traditionally recognized for treatment of cerebrovascular diseases. To evaluate the potential clinical use of the fruits of Toona sinensis Roem, we determined the dose dependence of the neuroprotective efficacy in a focal cerebral ischemic reperfusion model of rats and explored the underlying mechanisms. MATERIALS AND METHODS: Rats were subjected to occlusion of the middle cerebral artery (MCAO) by a nylon filament and treated with different doses (20mg/kg and 30 mg/kg) of n-butanol soluble fraction of the water extract of Chinese toon fruit or the vehicle for 1 week before induction of ischemia, s.i.d.. RESULTS: n-Butanol soluble fraction of the water extract of Chinese toon fruit reduced in a dose-dependent manner the ischemia-induced cerebral infarct and edema volume and attenuated neurological deficits observed at 6h point after ischemia. n-Butanol soluble fraction of the water extract of Chinese toon fruit reduced the levels of nitrate, nitrite, lipid peroxidation, cyclooxygenase-1, thromboxane in post-ischemic brain. n-Butanol soluble fraction of the water extract of Chinese toon fruit adjusted the elevation of the activity of glutathione peroxidase and superoxide dismutase in ischemic brain. CONCLUSIONS: The present study was the first evidence of effectiveness of n-butanol soluble fraction of the water extract of Chinese toon fruit in the rat stroke models, as it reduced infarct volume, inhibited the oxidative stress and inflammation.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Frutas/química , Inflamação/tratamento farmacológico , Meliaceae/química , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , 1-Butanol/química , Animais , Epoprostenol/genética , Epoprostenol/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/metabolismo , Inflamação/metabolismo , Masculino , Malondialdeído/metabolismo , Extratos Vegetais/química , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/metabolismo , Tromboxano A2/genética , Tromboxano A2/metabolismo , Água/química
15.
Circulation ; 128(9): 982-94, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23841984

RESUMO

BACKGROUND: Intravenous prostacyclin is approved for treating pulmonary arterial hypertension (PAH), but it has a short half-life and must be delivered systemically via an indwelling intravenous catheter. We hypothesize that localized jugular vein delivery of prostacyclin-producing cells may provide sustained therapeutic effects without the limitations of systemic delivery. METHODS AND RESULTS: We generated a vector expressing a human cyclooxygenase isoform 1 and prostacyclin synthase fusion protein that produces prostacyclin from arachidonic acid. Endothelial-like progenitor cells (ELPCs) were transfected with the cyclooxygenase isoform 1-prostacyclin synthase plasmid and labeled with lentivirus expressing nuclear-localized red fluorescent protein (nuRFP). The engineered ELPCs (expressing cyclooxygenase isoform 1-prostacyclin synthase and nuRFP) were tested in rats with monocrotaline (MCT)-induced PAH. In PAH prevention studies, treatment with engineered ELPCs or control ELPCs (expressing nuRFP alone) attenuated MCT-induced right ventricular systolic pressure increase, right ventricular hypertrophy, and pulmonary vessel wall thickening. Engineered ELPCs were more effective than control ELPCs in all variables evaluated. In PAH reversal studies, engineered ELPCs or control ELPCs increased the survival rate of rats with established PAH and decreased right ventricular hypertrophy. Engineered ELPCs provided a survival benefit 2 weeks earlier than did control ELPCs. Microarray-based gene ontology analysis of the right ventricle revealed that a number of MCT-altered genes and neurotransmitter pathways (dopamine, serotonin, and γ-aminobutyric acid) were restored after ELPC-based prostacyclin gene therapy. CONCLUSIONS: Cyclooxygenase isoform 1-prostacyclin synthase-expressing ELPCs reversed MCT-induced PAH. A single jugular vein injection offered survival benefits for at least 4 weeks and may provide a promising option for PAH patients.


Assuntos
Células Endoteliais/transplante , Epoprostenol/genética , Terapia Genética , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/terapia , Hipertrofia Ventricular Direita/terapia , Monocrotalina/efeitos adversos , Transplante de Células-Tronco , Animais , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Modelos Animais de Doenças , Epoprostenol/metabolismo , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita/mortalidade , Hipertrofia Ventricular Direita/patologia , Infusões Intravenosas , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Masculino , Ratos , Ratos Endogâmicos F344 , Taxa de Sobrevida , Engenharia Tecidual , Transfecção , Resultado do Tratamento
16.
Int J Oncol ; 43(3): 746-54, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23807031

RESUMO

Prostacyclin synthase (PGIS or PTGIS) is an enzyme that catalyses the conversion of prostaglandin H2 (PGH2) to prostaglandin I2 (PGI2). PGI2 promotes cancer growth by activating peroxisome proliferator-activated receptor δ (PPARδ), and increases the expression levels of the pro-angiogenic factor vascular endothelial growth factor (VEGF). We found that the expression of the PGIS gene was enhanced in WI-38, TIG-3-20 and HEL human lung fibroblast cells and two cancer cell lines (NB-1 and G361) under hypoxic conditions. The main localization of PGIS changed from the cytoplasm to the nucleus by hypoxia in WI-38 cells. The induced PGIS had an enzymatic activity since the intracellular level of 6-keto-prostaglandin, a useful marker of PGI2 biosynthesis in vivo, was increased with the increasing levels of PGIS. Expression of VEGF was increased in parallel with PGIS induction under hypoxic conditions. PGIS knockdown resulted in the decreased expression of VEGF mRNA. Since VEGF is a known PPARδ target gene, we examined the effects of siRNAs targeting PPARδ on the expression of VEGF under hypoxic conditions. Knockdown of PPARδ suppressed the expression of VEGF under hypoxic conditions in WI-38 cells. These findings suggest that PGIS is induced by hypoxia and regulates the expression of VEGF in fibroblasts. Fibroblasts in the hypoxic area of tumors may have an important role in tumor growth and angiogenesis.


Assuntos
Hipóxia Celular/genética , Sistema Enzimático do Citocromo P-450/genética , Fibroblastos/metabolismo , Oxirredutases Intramoleculares/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Linhagem Celular Tumoral , Sistema Enzimático do Citocromo P-450/biossíntese , Epoprostenol/genética , Epoprostenol/metabolismo , Fibroblastos/patologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Oxirredutases Intramoleculares/biossíntese , Pulmão/citologia , Pulmão/metabolismo , PPAR gama/genética , Prostaglandina H2/genética , Prostaglandina H2/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
18.
Eur J Pharmacol ; 700(1-3): 65-73, 2013 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23276663

RESUMO

Statins are reported to alleviate renal fibrosis in animal models with ureteral obstruction. However, the molecular mechanism of this antifibrotic effect is still unclear. Pressure force is an important mechanism contributing to induction and progression of tubulointerstitial fibrogenesis in ureteric obstruction. In this study, we investigated the influence of rosuvastatin on pressure-induced fibrotic responses in rat renal tubular cells (NRK-52E). We established an in vitro pressure culture system to study pressure-induced fibrotic responses in NRK-52E cells. When NRK-52E cells were cultured in the pressure culture system, 60 mm Hg of pressure induced the expression of connective tissue growth factor (CTGF), transforming growth factor (TGF)-ß, fibronectin, Smad3, and phospho-Smad3. Rosuvastatin significantly reduced these pressure-induced fibrotic responses at concentrations above 10 µM. Rosuvastatin also reduced the TGF-ß-induced expression of fibronectin and CTGF in NRK-52E cells. Pretreatment with rosuvastatin significantly induced prostacyclin (PGI(2)) generation, but reduced pressure-induced prostaglandin E(2) (PGE(2)). PGI(2) synthase small interfering RNA (siRNA) transfection significantly inhibited rosuvastatin-induced peroxisome proliferator-activated receptor α activation. The blockage of peroxisome proliferator-activated receptor α by siRNA transfection reduced the inhibitory effect of rosuvastatin on pressure-induced fibrotic responses. N-[2-(cyclohexyloxy)-4-nitrophenyl]-methanesulfonamide (NS398), a specific inhibitor of cyclooxygenase-2, diminished pressure-induced PGE(2) generation, and also reduced pressure-induced fibrotic responses. Additionally, PGE(2) decreased the antifibrotic effect of rosuvastatin. In conclusion, rosuvastatin reduces pressure-induced fibrotic responses in renal tubular cells by enhancing the PGI(2)-peroxisome proliferator-activated receptor α pathway and reducing PGE(2) generation.


Assuntos
Dinoprostona/metabolismo , Epoprostenol/metabolismo , Fluorbenzenos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Túbulos Renais/patologia , Pressão/efeitos adversos , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Animais , Linhagem Celular , Dinoprostona/biossíntese , Dinoprostona/genética , Epoprostenol/biossíntese , Epoprostenol/genética , Fibrose , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Ratos , Rosuvastatina Cálcica
19.
Circ J ; 77(4): 1053-62, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23257313

RESUMO

BACKGROUND: Prostacyclin (PGI2) enhances angiogenesis, especially in cooperation with bone marrow (BM)-derived endothelial progenitor cells (EPCs). However, the mechanisms of PGI2 in EPC-mediated angiogenesis in vivo remain unclear. The purpose of this study was to clarify the role of PGI2 in EPC-mediated angiogenesis using BM-specific IP deletion mice. METHODS AND RESULTS: Hind limb ischemia (HLI) was induced in wild-type (WT) mice transplanted with IP-deleted BM (WT/BM(IP(-/-)). Recovery of blood flow (RBF) in WT/BM(IP(-/-)) was impaired for 28 days after HLI, whereas RBF in IP(-/-)/BM(WT) was attenuated for up to 7 days compared with WT/BM(WT). The impaired RBF in WT/BM(IP(-/-)) was completely recovered by intramuscular injection of WT EPCs but not IP(-/-) EPCs. The impaired effects of IP(-/-) EPCs were in accordance with reduced formation of capillary and arterioles in ischemic muscle. An ex vivo aortic ring assay revealed that microvessel formation was enhanced by accumulation/adhesion of EPCs to perivascular sites as pericytes. IP(-/-)EPCs, in which expression of integrins was decreased, had impaired production of angiogenic cytokines, adhesion to neovessels and their angiogenic effects. The small-interfering RNA (siRNA)-mediated knockdown of integrin ß1 in WT EPCs attenuated adhesion to microvessels and their in vivo and in vitro angiogenic effects. CONCLUSIONS: PGI2 may induce persistent angiogenic effects in HLI through adhesion of EPCs to perivascular sites of neovessels via integrins in addition to paracrine effects.


Assuntos
Transplante de Medula Óssea , Células Endoteliais/metabolismo , Epoprostenol/metabolismo , Isquemia/terapia , Microcirculação , Neovascularização Fisiológica , Células-Tronco/metabolismo , Animais , Adesão Celular , Modelos Animais de Doenças , Células Endoteliais/patologia , Epoprostenol/genética , Membro Posterior/irrigação sanguínea , Membro Posterior/metabolismo , Membro Posterior/patologia , Isquemia/genética , Isquemia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pericitos/metabolismo , Pericitos/patologia , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Células-Tronco/patologia
20.
Theriogenology ; 78(9): 2071-86, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23043950

RESUMO

Prostaglandins (PGs) are critical regulators of a number of reproductive processes, including embryo development and implantation. In the present study, prostacyclin (PGI(2)) synthase (PGIS) mRNA and protein expression, as well as 6-keto PGF(1α) (a PGI(2) metabolite) concentration, were investigated in the pig uterus. Endometrial tissue and uterine luminal flushings were obtained on Days 4 to 18 of the estrous cycle and pregnancy. Additionally, conceptuses were collected and examined for PGIS mRNA expression and 6-keto PGF(1α) concentration. Regulation of PGI(2) synthesis in the porcine endometrium by steroids, conceptus products, and cytokines was studied in vitro and/or in vivo. Endometrial PGIS protein level increased on Days 12 and 16 in pregnant but not in cyclic gilts. Moreover, higher PGIS protein expression on Day 12 of pregnancy was accompanied by a greater content of 6-keto PGF(1α) in the endometrium. The concentration of 6-keto PGF(1α) in uterine luminal flushings increased substantially on Days 16 and 18 in pregnant gilts and was higher than in cyclic animals. Greater PGIS mRNA expression and PGI(2) metabolite concentration were detected in Day 12 and 14 conceptuses, respectively. Incubation of endometrial explants with conceptus-conditioned medium resulted in upregulation of PGIS protein expression and increased PGI(2) secretion. Moreover, PGIS mRNA and protein expression were upregulated in the endometrium collected from gravid uterine horn on Day 14 of pregnancy. In summary, PGIS is differentially expressed in the endometrium of cyclic and pregnant gilts resulting in higher PGI(2) synthesis in pregnant animals. Porcine conceptuses are important regulators of endometrial PGIS expression and PGI(2) release during the implantation period.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Endométrio/metabolismo , Epoprostenol/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Oxirredutases Intramoleculares/metabolismo , Suínos/fisiologia , 6-Cetoprostaglandina F1 alfa/genética , 6-Cetoprostaglandina F1 alfa/metabolismo , Animais , Western Blotting , Sistema Enzimático do Citocromo P-450/genética , Epoprostenol/genética , Feminino , Técnicas Imunoenzimáticas/veterinária , Oxirredutases Intramoleculares/genética , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA