Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
J Intern Med ; 296(1): 39-52, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38704820

RESUMO

Platelet hyperreactivity and hyperlipidaemia contribute significantly to atherosclerosis. Thus, it is desirable to review the platelet-hyperlipidaemia interplay and its impact on atherogenesis. Native low-density lipoprotein (nLDL) and oxidized LDL (oxLDL) are the key proatherosclerotic components of hyperlipidaemia. nLDL binds to the platelet-specific LDL receptor (LDLR) ApoE-R2', whereas oxLDL binds to the platelet-expressed scavenger receptor CD36, lectin-type oxidized LDLR 1 and scavenger receptor class A 1. Ligation of nLDL/oxLDL induces mild platelet activation and may prime platelets for other platelet agonists. Platelets, in turn, can modulate lipoprotein metabolisms. Platelets contribute to LDL oxidation by enhancing the production of reactive oxygen species and LDLR degradation via proprotein convertase subtilisin/kexin type 9 release. Platelet-released platelet factor 4 and transforming growth factor ß modulate LDL uptake and foam cell formation. Thus, platelet dysfunction and hyperlipidaemia work in concert to aggravate atherogenesis. Hypolipidemic drugs modulate platelet function, whereas antiplatelet drugs influence lipid metabolism. The research prospects of the platelet-hyperlipidaemia interplay in atherosclerosis are also discussed.


Assuntos
Aterosclerose , Plaquetas , Hiperlipidemias , Lipoproteínas LDL , Humanos , Aterosclerose/etiologia , Plaquetas/metabolismo , Lipoproteínas LDL/metabolismo , Ativação Plaquetária/fisiologia , Receptores de LDL/metabolismo , Hipolipemiantes/uso terapêutico
2.
J Transl Med ; 22(1): 419, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702818

RESUMO

BACKGROUND: Glioblastoma is an aggressive brain tumor linked to significant angiogenesis and poor prognosis. Anti-angiogenic therapies with vascular endothelial growth factor receptor 2 (VEGFR2) inhibition have been investigated as an alternative glioblastoma treatment. However, little is known about the effect of VEGFR2 blockade on glioblastoma cells per se. METHODS: VEGFR2 expression data in glioma patients were retrieved from the public database TCGA. VEGFR2 intervention was implemented by using its selective inhibitor Ki8751 or shRNA. Mitochondrial biogenesis of glioblastoma cells was assessed by immunofluorescence imaging, mass spectrometry, and western blot analysis. RESULTS: VEGFR2 expression was higher in glioma patients with higher malignancy (grade III and IV). VEGFR2 inhibition hampered glioblastoma cell proliferation and induced cell apoptosis. Mass spectrometry and immunofluorescence imaging showed that the anti-glioblastoma effects of VEGFR2 blockade involved mitochondrial biogenesis, as evidenced by the increases of mitochondrial protein expression, mitochondria mass, mitochondrial oxidative phosphorylation (OXPHOS), and reactive oxygen species (ROS) production, all of which play important roles in tumor cell apoptosis, growth inhibition, cell cycle arrest and cell senescence. Furthermore, VEGFR2 inhibition exaggerated mitochondrial biogenesis by decreased phosphorylation of AKT and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), which mobilized PGC1α into the nucleus, increased mitochondrial transcription factor A (TFAM) expression, and subsequently enhanced mitochondrial biogenesis. CONCLUSIONS: VEGFR2 blockade inhibits glioblastoma progression via AKT-PGC1α-TFAM-mitochondria biogenesis signaling cascade, suggesting that VEGFR2 intervention might bring additive therapeutic values to anti-glioblastoma therapy.


Assuntos
Apoptose , Proliferação de Células , Glioblastoma , Mitocôndrias , Biogênese de Organelas , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Humanos , Glioblastoma/patologia , Glioblastoma/metabolismo , Glioblastoma/tratamento farmacológico , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proliferação de Células/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Linhagem Celular Tumoral , Apoptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Cardiovasc Diabetol ; 23(1): 150, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702777

RESUMO

BACKGROUND: Vasculopathy is the most common complication of diabetes. Endothelial cells located in the innermost layer of blood vessels are constantly affected by blood flow or vascular components; thus, their mechanosensitivity plays an important role in mediating vascular regulation. Endothelial damage, one of the main causes of hyperglycemic vascular complications, has been extensively studied. However, the role of mechanosensitive signaling in hyperglycemic endothelial damage remains unclear. METHODS: Vascular endothelial-specific Piezo1 knockout mice were generated to investigate the effects of Piezo1 on Streptozotocin-induced hyperglycemia and vascular endothelial injury. In vitro activation or knockdown of Piezo1 was performed to evaluate the effects on the proliferation, migration, and tubular function of human umbilical vein endothelial cells in high glucose. Reactive oxygen species production, mitochondrial membrane potential alternations, and oxidative stress-related products were used to assess the extent of oxidative stress damage caused by Piezo1 activation. RESULTS: Our study found that in VECreERT2;Piezo1flox/flox mice with Piezo1 conditional knockout in vascular endothelial cells, Piezo1 deficiency alleviated streptozotocin-induced hyperglycemia with reduced apoptosis and abscission of thoracic aortic endothelial cells, and decreased the inflammatory response of aortic tissue caused by high glucose. Moreover, the knockout of Piezo1 showed a thinner thoracic aortic wall, reduced tunica media damage, and increased endothelial nitric oxide synthase expression in transgenic mice, indicating the relief of endothelial damage caused by hyperglycemia. We also showed that Piezo1 activation aggravated oxidative stress injury and resulted in severe dysfunction through the Ca2+-induced CaMKII-Nrf2 axis in human umbilical vein endothelial cells. In Piezo1 conditional knockout mice, Piezo1 deficiency partially restored superoxide dismutase activity and reduced malondialdehyde content in the thoracic aorta. Mechanistically, Piezo1 deficiency decreased CaMKII phosphorylation and restored the expression of Nrf2 and its downstream molecules HO-1 and NQO1. CONCLUSION: In summary, our study revealed that Piezo1 is involved in high glucose-induced oxidative stress injury and aggravated endothelial dysfunction, which have great significance for alleviating endothelial damage caused by hyperglycemia.


Assuntos
Glicemia , Diabetes Mellitus Experimental , Células Endoteliais da Veia Umbilical Humana , Canais Iônicos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III , Estresse Oxidativo , Animais , Humanos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Diabetes Mellitus Experimental/metabolismo , Canais Iônicos/metabolismo , Canais Iônicos/genética , Glicemia/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Mecanotransdução Celular , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/deficiência , Células Cultivadas , Proliferação de Células , Apoptose , Masculino , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/fisiopatologia , Angiopatias Diabéticas/patologia , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/etiologia , Movimento Celular , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Camundongos , Estreptozocina , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Endotélio Vascular/patologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética
4.
Cell Mol Life Sci ; 79(5): 247, 2022 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-35437611

RESUMO

BACKGROUND AND AIM: Platelets are an able regulator of CD4+ T cell immunity. Herein, the mechanisms underlying platelet-regulated effector responses of naïve CD4+ T (Tn) cells were investigated. METHODS: Platelet-Tn cell co-cultures of human cells, genetically modified murine models, and high-throughput bioinformatic analyses were combined to elucidate molecular mechanisms of platelet-dependent regulation. RESULTS: Platelets exerted sophisticated regulation on effector responses of type 1, 2, and 17 T helper (Th1/Th2/Th17) and regulatory T (Treg) cells, in time-, concentration-, and organ-dependent manners and with close cooperation of transforming growth factor ß (TGFß) and platelet factor 4 (PF4). PF4 at low concentrations reinforced TGFß signaling by heteromerizing with type III TGFß receptor (TGFBRIII), and subsequently enhanced TGFBRII expression and TGFß signaling. High-concentration PF4 had, however, opposite effects by directly binding to TGFBRII, blocking TGFß-TGFBRII ligation, and thus inhibiting TGFß signaling. Furthermore, platelet depletion markedly hampered Treg and Th17 responses in the spleen but not in the lymph nodes, blockade of platelet-Tn cell contact diminished platelet effects, while spleen injection of PF4-immobilized microparticles in PF4-deficient mice mimicked platelet effects, suggesting the importance of direct platelet-Tn contact and platelet-bound PF4 for the optimal regulatory effects by platelets. CONCLUSION: Platelets exert context-dependent regulations on effector responses of Tn cells via PF4-TGFß duet, suggesting new possibilities of platelet-targeted interventions of T cell immunity.


Assuntos
Fator Plaquetário 4 , Fator de Crescimento Transformador beta , Animais , Plaquetas/metabolismo , Linfócitos T CD4-Positivos , Camundongos , Fator Plaquetário 4/metabolismo , Linfócitos T Reguladores , Fator de Crescimento Transformador beta/metabolismo
5.
Platelets ; 33(3): 360-370, 2022 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-34137652

RESUMO

Platelets regulate multiple aspects of CD4+ T cell immunity, and may exert distinct regulations among different T cell subsets. Our aim was to investigate how platelets regulate CD4+ central memory T cell (Tcm) responses. αCD3/αCD28-stimulated human CD4+ Tcm cells were cultured without or with platelets or platelet-derived mediators. Polyclonal stimulation induced cell proliferation and Th1 and Treg cell activation of Tcm cells. Platelet factor 4/PF4 neutralization abolished platelet-enhanced Tcm effector responses, whilst TGFß neutralization only partially inhibited platelet-enhanced Treg cell activation. PF4 supplementation mimicked the effects of platelet co-cultures, while PF4 receptor CXCR3 blockade and CXCR3 knockdown with siRNAs inhibited or abolished PF4-enhanced Th1 and Treg cell responses. Platelet co-cultures or PF4-treatment increased Tcm cell proliferation, whilst CXCR3 blockade counteracted. PF4-enhanced Tcm proliferation and effector cell responses were associated with mitochondrial biogenesis. Overexpression of mitochondrial transcription factor A (TFAM) mimicked PF4 effects, and PF4 treatment attenuated Akt phosphorylation of activated Tcm cells, leading to mitochondrial biogenesis. Impacts of platelets and PF4 on Tcm proliferation were further confirmed by that CXCR3 knockdown/blockade counteracted PF4-enhanced Tcm cell proliferation. In conclusion, platelets enhance Th1 and Treg cell responses of CD4+ Tcm cells, via PF4-dependent mitochondrial biogenesis and cell proliferation of Tcm cells.


Assuntos
Plaquetas/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células T de Memória/metabolismo , Fator Plaquetário 4/imunologia , Adulto , Proliferação de Células , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Biogênese de Organelas , Adulto Jovem
6.
J Cell Mol Med ; 25(1): 434-447, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33225628

RESUMO

Alzheimer's disease (AD) is the most common dementia, characterized by pathological accumulation of ß-amyloid (Aß) and hyperphosphorylation of tau protein, together with a damaging chronic inflammation. The lack of effective treatments urgently warrants new therapeutic strategies. Resolution of inflammation, associated with beneficial and regenerative activities, is mediated by specialized pro-resolving lipid mediators (SPMs) including maresin 1 (MaR1). Decreased levels of MaR1 have been observed in AD brains. However, the pro-resolving role of MaR1 in AD has not been fully investigated. In the present study, human monocyte-derived microglia (MdM) and a differentiated human monocyte cell line (THP-1 cells) exposed to Aß were used as models of AD neuroinflammation. We have studied the potential of MaR1 to inhibit pro-inflammatory activation of Aß and assessed its ability to stimulate phagocytosis of Aß42 . MaR1 inhibited the Aß42 -induced increase in cytokine secretion and stimulated the uptake of Aß42 in both MdM and differentiated THP-1 cells. MaR1 was also found to decrease chemokine secretion and reduce the associated increase in the activation marker CD40. Activation of kinases involved in transduction of inflammation was not affected by MaR1, but the activity of nuclear factor (NF)-κB was decreased. Our data show that MaR1 exerts effects that indicate a pro-resolving role in the context of AD and thus presents itself as a potential therapeutic target for AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Microglia/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Western Blotting , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Ácidos Docosa-Hexaenoicos/genética , Humanos , Imuno-Histoquímica , NF-kappa B/metabolismo , Fagocitose/genética , Fagocitose/fisiologia , Células THP-1
7.
Angiogenesis ; 23(4): 515-529, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32451774

RESUMO

Angiogenesis is critical for the initiation and progression of solid tumors, as well as hematological malignancies. While angiogenesis in solid tumors has been well characterized, a large body of investigation is devoted to clarify the impact of angiogenesis on lymphoma development. B-cell non-Hodgkin lymphoma (B-NHL) is the most common lymphoid malignancy with a highly heterogeneity. The malignancy remains incurable despite that the addition of rituximab to conventional chemotherapies provides substantial improvements. Several angiogenesis-related parameters, such as proangiogenic factors, circulating endothelial cells, microvessel density, and tumor microenvironment, have been identified as prognostic indicators in different types of B-NHL. A better understanding of how these factors work together to facilitate lymphoma-specific angiogenesis will help to design better antiangiogenic strategies. So far, VEGF-A monoclonal antibodies, receptor tyrosine kinase inhibitors targeting VEGF receptors, and immunomodulatory drugs with antiangiogenic activities are being tested in preclinical and clinical studies. This review summarizes recent advances in the understanding of the role of angiogenesis in B-NHL, and discusses the applications of antiangiogenic therapies.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Linfócitos B/imunologia , Linfoma não Hodgkin/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Animais , Linfócitos B/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Humanos , Microambiente Tumoral/efeitos dos fármacos
8.
Platelets ; 31(6): 777-783, 2020 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-31610683

RESUMO

INTRODUCTION: Platelets are critical for hemostasis, and a low platelet count predicts mortality in trauma. The role of platelet dysfunction in severe traumatic hemorrhage and coagulopathy needs to be further defined. The aim of this study was to evaluate the platelet function in a new model of experimental traumatic hemorrhage. MATERIAL AND METHODS: New Zealand white rabbits (n = 10) were subjected to tracheostomy and trauma laparotomy, and then bilateral femur fractures with 40% hemorrhage of their estimated blood volume. Arterial blood gases, standard coagulation tests, mean platelet volume, platelet aggregation using impedance aggregometry with agonist collagen, arachidonic acid (ASPI), and adenosine diphosphate (ADP), rotational thromboelastometry, and fibrinogen binding of platelets were analyzed using flow cytometry. RESULTS: After traumatic hemorrhage, there was a significant physiological response with a rise in lactate (P < .001) and a decrease in base excess (P < .001) and temperature (P < .001). Platelet count decreased from a mean of 244x109/L to 94 x109/L (P = .004) and the mean platelet volume increased from 5.1fL to 6.1fL (P = .002). Impedance aggregometry with the agonist collagen, ASPI, and ADP was all significantly decreased after hemorrhage (P = .007). However, there was an increased fibrinogen binding of ADP-activated platelets after traumatic hemorrhage analyzed by flow cytometry (P < .05). CONCLUSIONS: This traumatic hemorrhage model presents two parallel pathophysiological responses of platelets; platelet consumption as evidenced by a significant decrease in platelet count and aggregation, and platelet hyperreactivity as shown by a higher mean platelet volume and enhanced platelet fibrinogen binding. Further studies are needed to characterize these different aspects of platelet function in severe traumatic hemorrhage.


Assuntos
Plaquetas/metabolismo , Hemorragia/sangue , Ferimentos e Lesões/sangue , Animais , Plaquetas/citologia , Modelos Animais de Doenças , Humanos , Coelhos
9.
Arterioscler Thromb Vasc Biol ; 38(5): 1115-1124, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29599139

RESUMO

OBJECTIVE: Deletion of mPGES-1 (microsomal prostaglandin E synthase-1)-an anti-inflammatory target alternative to COX (cyclooxygenase)-2-attenuates injury-induced neointima formation in mice. This is attributable to the augmented levels of PGI2 (prostacyclin)-a known restraint of the vascular response to injury, acting via IP (I prostanoid receptor). To examine the role of mPGES-1-derived PGE2 (prostaglandin E2) in vascular remodeling without the IP. APPROACH AND RESULTS: Mice deficient in both IP and mPGES-1 (DKO [double knockout] and littermate controls [IP KO (knockout)]) were subjected to angioplasty wire injury. Compared with the deletion of IP alone, coincident deletion of IP and mPGES-1 increased neointima formation, without affecting media area. Early pathological changes include impaired reendothelialization and increased leukocyte invasion in neointima. Endothelial cells (ECs), but not vascular smooth muscle cells, isolated from DKOs exhibited impaired cell proliferation. Activation of EP (E prostanoid receptor) 4 (and EP2, to a lesser extent), but not of EP1 or EP3, promoted EC proliferation. EP4 antagonism inhibited proliferation of mPGES-1-competent ECs, but not of mPGES-1-deficient ECs, which showed suppressed PGE2 production. EP4 activation inhibited leukocyte adhesion to ECs in vitro, promoted reendothelialization, and limited neointima formation post-injury in the mouse. Endothelium-restricted deletion of EP4 in mice suppressed reendothelialization, increased neointimal leukocytes, and exacerbated neointimal formation. CONCLUSIONS: Removal of the IP receptors unmasks a protective role of mPGES-1-derived PGE2 in limiting injury-induced vascular hyperplasia. EP4, in the endothelial compartment, is essential to promote reendothelialization and restrain neointimal formation after injury. Activating EP4 bears therapeutic potential to prevent restenosis after percutaneous coronary intervention.


Assuntos
Proliferação de Células , Dinoprostona/metabolismo , Células Endoteliais/enzimologia , Artéria Femoral/enzimologia , Prostaglandina-E Sintases/metabolismo , Receptores de Epoprostenol/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Lesões do Sistema Vascular/enzimologia , Animais , Adesão Celular , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Feminino , Artéria Femoral/lesões , Artéria Femoral/patologia , Humanos , Leucócitos/metabolismo , Leucócitos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso/enzimologia , Músculo Liso/patologia , Neointima , Prostaglandina-E Sintases/deficiência , Prostaglandina-E Sintases/genética , Reepitelização , Receptores de Epoprostenol/deficiência , Receptores de Epoprostenol/genética , Receptores de Prostaglandina E Subtipo EP4/deficiência , Receptores de Prostaglandina E Subtipo EP4/genética , Transdução de Sinais , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia
10.
J Cell Mol Med ; 22(2): 808-822, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29063670

RESUMO

Emerging evidence indicates that irisin provides beneficial effects in diabetes. However, whether irisin influences the development of diabetic cardiomyopathy (DCM) remains unclear. Therefore, we investigated the potential role and mechanism of action of irisin in diabetes-induced myocardial dysfunction in mice. Type 1 diabetes was induced in mice by injecting streptozotocin, and the diabetic mice were administered recombinant r-irisin (low or high dose: 0.5 or 1.5 µg/g body weight/day, I.P.) or PBS for 16 weeks. Irisin treatment did not alter blood glucose levels in the diabetic mice. However, the results of echocardiographical and histopathological assays indicated that low-dose irisin treatment alleviated cardiac fibrosis and left ventricular function in the diabetic mice, whereas high-dose irisin failed to mitigate the ventricular function impairment and increased collagen deposition. The potential mechanism underlying the effect of low-dose irisin involved irisin-mediated inhibition of high glucose-induced endothelial-to-mesenchymal transition (EndMT); conversely, high-dose irisin treatment enhanced high glucose-induced MMP expression by stimulating MAPK (p38 and ERK) signalling and cardiac fibroblast proliferation and migration. Low-dose irisin alleviated DCM development by inhibiting high glucose-induced EndMT. By contrast, high-dose irisin disrupted normal MMP expression and induced cardiac fibroblast proliferation and migration, which results in excess collagen deposition. Thus, irisin can inhibit high glucose-induced EndMT and exert a dose-dependent bidirectional effect on DCM.


Assuntos
Cardiomiopatias Diabéticas/patologia , Fibronectinas/farmacologia , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/patologia , Mesoderma/patologia , Animais , Glicemia/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/fisiopatologia , Cardiomiopatias Diabéticas/sangue , Cardiomiopatias Diabéticas/fisiopatologia , Ativação Enzimática/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Mesoderma/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Estreptozocina , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
BMC Cancer ; 18(1): 515, 2018 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-29720123

RESUMO

BACKGROUND: Gastric cancer is common in developing regions, and we hope to find out an economical but practical prognostic indicator. It was reported that pre-treatment peripheral neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR), as well as differentiation status, were associated with cancer progression. Hence, we introduced a novel combined Neutrophil/platelet/lymphocyte/differentiation Score (cNPLDS) to improve the prediction value of palliative chemotherapeutic response in advanced gastric cancer. METHODS: According to statistical sample size estimation, 136 primary diagnosed unresectable advanced ptaients were included for a retrospective study. The follow-up end-point was progression free survival (PFS) during the first-line palliative chemotherapy. Differentiation stratified patients into well, medium and poor groups by score 1 to 3, while patients with neither elevated NLR and PLR, only one elevated, or both elevated were of the combined NLR-PLR score (cNPS) 1 to 3, respectively. The cNPLDS was calculated by multiplying the tumor differentiation score and cNPS. RESULTS: Determined by the receiver operating characteristic (ROC) curve, the optimal cut-off points for NLR and PLR were 3.04 and 223. Through univariate analysis and survival analysis, poor differentiation, high NLR, high PLR, high cNPS, and high cNPLDS respectively indicated inferior PFS during the first-line palliative chemotherapy. Patients were furhter classified into low to high risk groups by cNPLDS. Groups of elevated NLR, PLR, cNPS, and cNPLDS showed lower disease control rate. Compared to other parameters, cNPLDS significantly improved the accuracy in predicing the first-progression. CONCLUSIONS: This study indicates that the novel parameter cNPLDS is superior to NLR or PLR alone, or even cNPS, in predicting the first-line chemosensitivity in advanced gastric cancer.


Assuntos
Plaquetas/imunologia , Linfócitos/imunologia , Neutrófilos/imunologia , Neoplasias Gástricas/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Área Sob a Curva , Biomarcadores Tumorais/análise , Plaquetas/patologia , Diferenciação Celular/imunologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Feminino , Humanos , Contagem de Linfócitos , Linfócitos/patologia , Masculino , Pessoa de Meia-Idade , Neutrófilos/patologia , Contagem de Plaquetas , Prognóstico , Intervalo Livre de Progressão , Curva ROC , Estudos Retrospectivos , Sensibilidade e Especificidade , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia
12.
Br J Cancer ; 117(5): 695-703, 2017 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-28697175

RESUMO

BACKGROUND: Selective platelet release of pro- or anti-angiogenic factors distinctly regulated angiogenesis. We hypothesised that selective release of platelet angiogenic factors could differently regulate tumour growth. METHODS: Breast cancer cell proliferation, cancer cell-induced endothelial tube formation in vitro, and tumour growth in vivo were studied in the presence of protease-activated receptor 1-stimulated platelet releasate (PAR1-PR; rich in pro-angiogenic factors) or PAR4-PR (rich in anti-angiogenic factors). RESULTS: The PAR1-PR and PAR4-PR supplementation (10%) similarly enhanced cell proliferation of MCF-7 and MDA-MB-231 breast cancer cells. The cancer cells triggered capillary-like tube formation of endothelial cells that was further enhanced by pro-angiogenic factor-rich PAR1-PR. The VEGF, but not SDF-1α, receptor blockade abolished PAR1-PR/PAR4-PR-enhanced cancer cell proliferation. Integrin blockade by RGDS had identical effects as VEGF inhibition. The Src and ERK inhibition diminished, whereas PI3K and PKC blockade abolished platelet releasate-enhanced cancer cell proliferation. Using a model of subcutaneous implantation of MDA-MB-231 cells in nude mice, PAR1-PR enhanced tumour growth more markedly than PAR4-PR, and seemed to achieve the exaggeration by promoting more profound tumour angiogenesis. CONCLUSIONS: Platelet releasate increases breast cancer cell proliferation through VEGF-integrin cooperative signalling. Pro-angiogenic factor-rich platelet releasate enhances cancer cell-induced angiogenesis more markedly, and thus exaggerates tumour growth in vivo.


Assuntos
Plaquetas/metabolismo , Neoplasias da Mama/metabolismo , Integrinas/metabolismo , Neovascularização Patológica/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Animais , Plaquetas/efeitos dos fármacos , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Proliferação de Células , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrinas/antagonistas & inibidores , Células MCF-7 , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Compostos de Fenilureia/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Quinolinas/farmacologia , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
13.
Int J Cancer ; 138(9): 2078-87, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26356352

RESUMO

Cancer progress is accompanied by platelet activation and thrombotic complications. Platelets are a dangerous alliance of cancer cells, and are a close engager in multiple processes of cancer metastasis. Platelet adhesion to cancer cells forms a protective cloak that helps cancer cells to escape immune surveillance and natural killer cell-mediated cytolysis. Platelets facilitate tethering and arrest of disseminated cancer cells in the vasculature, enhance invasive potentials and thus extravasation of cancer cells. Moreover, platelets recruit monocytes and granulocytes to the sites of cancer cell arrest, and collaborate with them to establish a pro-metastatic microenvironment and metastatic niches. Platelets also secret a number of growth factors to stimulate cancer cell proliferation, release various angiogenic regulators to regulate tumor angiogenesis and subsequently promote cancer growth and progress. Albeit platelets are helping the "villain" cancer to do evil, the close engagements of platelets in cancer metastasis and progress can be used as the intervention targets for new anti-cancer therapeutic developments. Platelet-targeted anti-cancer strategy may bring in novel anti-cancer treatments that can synergize the therapeutic effects of chemotherapies and surgical treatments of cancer.


Assuntos
Plaquetas/patologia , Invasividade Neoplásica/patologia , Neoplasias/patologia , Animais , Humanos , Evasão Tumoral/fisiologia
14.
Int J Cancer ; 139(11): 2553-69, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27529686

RESUMO

Voltage-gated sodium channels (VGSCs), which are aberrantly expressed in several human cancers, affect cancer cell behavior; however, their role in gastric cancer (GC) and the link between these channels and tumorigenic signaling remain unclear. The aims of this study were to determine the clinicopathological significance and role of the VGSC Nav 1.7 in GC progression and to investigate the associated mechanisms. Here, we report that the SCN9A gene encoding Nav 1.7 was the most abundantly expressed VGSC subtype in GC tissue samples and two GC cell lines (BGC-823 and MKN-28 cells). SCN9A expression levels were also frequently found to be elevated in GC samples compared to nonmalignant tissues by real-time PCR. In the 319 GC specimens evaluated by immunohistochemistry, Nav 1.7 expression was correlated with prognosis, and transporter Na(+) /H(+) exchanger-1 (NHE1) and oncoprotein metastasis-associated in colon cancer-1 (MACC1) expression. Nav 1.7 suppression resulted in reduced voltage-gated sodium currents, decreased NHE1 expression, increased extracellular pH and decreased intracellular pH, and ultimately, reduced invasion and proliferation rates of GC cells and growth of GC xenografts in nude mice. Nav 1.7 inhibition led to reduced MACC1 expression, while MACC1 inhibition resulted in reduced NHE1 expression in vitro and in vivo. Mechanistically, the suppression of Nav 1.7 decreased NF-κB p65 nuclear translocation via p38 activation, thus reducing MACC1 expression. Downregulation of MACC1 decreased c-Jun phosphorylation and subsequently reduced NHE1 expression, whereas the addition of hepatocyte growth factor (HGF), a c-Met physiological ligand, reversed the effect. These results indicate that Nav 1.7 promotes GC progression through MACC1-mediated upregulation of NHE1. Therefore, Nav 1.7 is a potential prognostic marker and/or therapeutic target for GC.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Neoplasias Gástricas/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Fator de Crescimento de Hepatócito/metabolismo , Xenoenxertos , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Nus , Canal de Sódio Disparado por Voltagem NAV1.7/biossíntese , Canal de Sódio Disparado por Voltagem NAV1.7/genética , NF-kappa B/metabolismo , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/biossíntese , Trocadores de Sódio-Hidrogênio/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Transativadores , Regulação para Cima
15.
Prostaglandins Other Lipid Mediat ; 121(Pt A): 122-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26115646

RESUMO

Classical Hodgkin lymphoma (cHL) has a unique pathological feature characterized by a minority of malignant Hodgkin Reed-Sternberg (H-RS) cells surrounded by numerous inflammatory cells. Cysteinyl-leukotrienes (CysLTs) are produced by eosinophils, macrophages and mast cells in the HL tumor microenvironment. In the present study we have explored the signal transduction pathways leading to leukotriene (LT) D4 induced expression of cytokines in the Hodgkin lymphoma cell line L1236 and KM-H2. Stimulation of L1236 and KM-H2 cells with LTD4 led to a concentration- and time-dependent increase at the transcriptional level of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-8, chemokine (C-C motif) ligand 3 (CCL3) and CCL4. The expression of several transcription factors was induced upon stimulation of Hodgkin cell lines with LTD4. Among these, EGR-1 was required for cytokine production. Inhibition of EGR-1 expression using shEGR-1 transduced by lentivirus led to suppression of the expression of TNF-α and IL-6. The effect of LTD4 on the expression of transcription factors and cytokines were also blocked by the specific CysLT1 receptor antagonist zafirlukast. These results demonstrate that EGR-1 plays a critical role in LTD4-induced cytokine transcription in Hodgkin cell lines.


Assuntos
Citocinas/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Doença de Hodgkin/patologia , Leucotrieno D4/farmacologia , Transcrição Gênica/efeitos dos fármacos , Linhagem Celular Tumoral , Proteína 1 de Resposta de Crescimento Precoce/deficiência , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Doença de Hodgkin/genética , Humanos , Receptores de Leucotrienos/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Blood ; 117(14): 3907-11, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21330475

RESUMO

The present study characterized platelet secretion and surface expression of proangiogenic stromal cell-derived factor-1α (SDF-1α) and vascular endothelial growth factor (VEGF) and antiangiogenic PF4 and endostatin on activation. The angiogenic factors presented in randomly distributed granules in resting platelets, which were peripherized on activation. Confocal and immunogold electron microscopy demonstrated that SDF-1α/CXCL12 and PF4/CXCL4 mostly present in different granules. Platelet activation induced marked SDF-1α and endostatin but mild PF4 or no VEGF surface expression. PAR1-activating peptide (PAR1-AP), adenosine diphosphate (via P2Y1/P2Y12), and glycoprotein VI-targeting collagen-related peptide induced massive SDF-1α and VEGF but modest PF4 or no endostatin release. In contrast, PAR4-AP triggered marked PF4 and sole endostatin release but limited SDF-1α or VEGF secretion. Distinct platelet release of SDF-1α and endostatin involved different engagements of intracellular signaling pathways. In conclusion, different platelet stimuli evoke distinct secretion and surface expression of proangiogenic and antiangiogenic factors. PAR1, adenosine diphosphate, and glycoprotein VI stimulation favors proangiogenic, whereas PAR4 promotes antiangiogenic, factor release.


Assuntos
Indutores da Angiogênese/metabolismo , Inibidores da Angiogênese/metabolismo , Plaquetas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ativação Plaquetária/fisiologia , Adulto , Plaquetas/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Quimiocina CXCL12/metabolismo , Endostatinas/metabolismo , Feminino , Imunofluorescência , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Peptídeos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptor PAR-1/agonistas , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Nat Commun ; 14(1): 7527, 2023 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-37980429

RESUMO

Mitochondria dysfunction contributes to acute liver injuries, and mitochondrial regulators, such as PGC-1α and MCJ, affect liver regeneration. Therefore, identification of mitochondrial modulators may pave the way for developing therapeutic strategies. Here, ZHX2 is identified as a mitochondrial regulator during acute liver injury. ZHX2 both transcriptionally inhibits expression of several mitochondrial electron transport chain genes and decreases PGC-1α stability, leading to reduction of mitochondrial mass and OXPHOS. Loss of Zhx2 promotes liver recovery by increasing mitochondrial OXPHOS in mice with partial hepatectomy or CCl4-induced liver injury, and inhibition of PGC-1α or electron transport chain abolishes these effects. Notably, ZHX2 expression is higher in liver tissues from patients with drug-induced liver injury and is negatively correlated with mitochondrial mass marker TOM20. Delivery of shRNA targeting Zhx2 effectively protects mice from CCl4-induced liver injury. Together, our data clarify ZHX2 as a negative regulator of mitochondrial OXPHOS and a potential target for developing strategies for improving liver recovery after acute injuries.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Fosforilação Oxidativa , Humanos , Camundongos , Animais , Doença Hepática Crônica Induzida por Substâncias e Drogas/metabolismo , Mitocôndrias/metabolismo , Hepatectomia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo
18.
J Cell Commun Signal ; 16(4): 661-675, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35414144

RESUMO

Platelets are well known for their roles in hemostasis and thrombosis, and are increasingly recognized for their abilities to interact with white blood cells during inflammatory diseases, via secreted soluble factors as well as cell-cell contact. This interaction has been investigated in animal models and patient samples and has shown to be implicated in patient outcomes in several diseases. Platelet-leukocyte co-cultures are widely used to study platelet-leukocyte interactions ex vivo. However, there is a paucity with regard to the systematic characterization of cell activation and functional behaviors of platelets and leukocytes in these co-cultures. Hence we aimed to characterize a model of platelet-leukocyte co-culture ex vivo. Human peripheral blood mononuclear cell (PBMC) and platelets were isolated and co-cultured for 5 days at 37 °C in the presence or absence of anti-CD3/CD28 antibodies or PHA. We evaluated PF-4 secretion and p-selectin expression in platelets as markers of platelet activation. Lymphocyte activation was assessed by cell proliferation and cell population phenotyping, in addition to platelet-lymphocyte aggregation. Platelet secretion and p-selectin expression is maintained throughout the co-culture, indicating that platelets were viable and reactive over the 5 days. Similarly PBMCs were viable and maintained proliferative capacity. Finally, dynamic heterotypic conjugation between platelets and T lymphocytes was also observed throughout co-culture (with a peak at days 3 and 4) upon T lymphocyte activation. In conclusion, this in vitro model can successfully mimic the in vivo interaction between platelets and T lymphocytes, and can be used to confirm and/or support in vivo results.

19.
Adv Sci (Weinh) ; 9(16): e2103135, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35398991

RESUMO

Covalently closed circular DNA (cccDNA) is the transcriptional template of hepatitis B virus (HBV), which interacts with both host and viral proteins to form minichromosome in the nucleus and is resistant to antiviral agents. Identification of host factors involved in cccDNA transcriptional regulation is expected to prove a new venue for HBV therapy. Recent evidence suggests the involvement of long noncoding RNAs (lncRNAs) in mediating the interaction of host factors with various viruses, however, lncRNAs that HBV targets and represses cccDNA transcription have not been fully elucidated. Here, the authors identified LINC01431 as a novel host restriction factor for HBV transcription. Mechanically, LINC01431 competitively bound with type I protein arginine methyltransferase (PRMT1) to block the HBx-mediated PRMT1 ubiquitination and degradation. Consequently, LINC01431 increased the occupancy of PRMT1 on cccDNA, leading to enhanced H4R3me2a modification and reduced acetylation of cccDNA-bound histones, thereby repressing cccDNA transcription. In turn, to facilitate viral replication, HBV transcriptionally repressed LINC01431 expression by HBx-mediated repression of transcription factor Zinc fingers and homeoboxes 2 (ZHX2). Collectively, the study demonstrates LINC01431 as a novel epigenetic regulator of cccDNA minichromosome and highlights a feedback loop of HBx-LINC01431-PRMT1 in HBV replication, which provides potential therapeutic targets for HBV treatment.


Assuntos
Vírus da Hepatite B , RNA Longo não Codificante , DNA Circular/genética , DNA Circular/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Vírus da Hepatite B/genética , Histonas/genética , Histonas/metabolismo , Metilação , RNA Longo não Codificante/genética
20.
Free Radic Biol Med ; 168: 95-109, 2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-33789125

RESUMO

The fundamental physiology of circulating red blood cells (RBCs) and platelets involving regulation of oxygen transport and hemostasis, respectively, are well-described in the literature. Their abundance in the circulation and their interaction with the vascular wall and each other have attracted the attention of other putative physiological and pathophysiological effects of these cells. RBCs and platelets are both important regulators of redox balance harboring powerful pro-oxidant and anti-oxidant (enzymatic and non-enzymatic) capacities. They are also involved in the regulation of vascular tone mainly via export of nitric oxide bioactivity and adenosine triphosphate. Of further importance are emerging observations that these cells undergo functional alterations when exposed to risk factors for cardiovascular disease and during developed cardiometabolic diseases. Under these conditions, the RBCs and platelets contribute to increased oxidative stress by their formation of reactive species including superoxide anion radical, hydrogen peroxide and peroxynitrite. These alterations trigger key changes in the vascular wall characterized by enhanced oxidative stress, reduced nitric oxide bioavailability and endothelial dysfunction. Additional pathophysiological effects are triggered in the heart resulting in increased susceptibility to ischemia-reperfusion injury with impairment in cardiac function. Pharmacological interventions aiming at restoring circulating cell function has been shown to exert marked beneficial effects on cardiovascular function. In this review, we summarize the current knowledge of RBC and platelet biology with special focus on redox biology, their roles in the development of cardiovascular disease and potential therapeutic strategies targeting RBC and platelet dysfunction. Finally, the complex and scarcely understood interaction between RBCs and platelets is discussed.


Assuntos
Plaquetas , Doenças Cardiovasculares , Eritrócitos , Humanos , Óxido Nítrico , Oxirredução , Estresse Oxidativo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA