Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 8723, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37253865

RESUMO

Antigen-naive IgM-producing B cells are atheroprotective, whereas mature B cells producing class-switched antibodies promote atherosclerosis. Activation-induced cytidine deaminase (AID), which mediates class switch recombination (CSR), would thus be expected to foster atherosclerosis. Yet, AID also plays a major role in the establishment of B cell tolerance. We sought to define whether AID affects atherosclerotic plaque formation. We generated Ldlr-/- chimeras transplanted with bone marrow from Aicda-/- or wild-type (WT) mice, fed a HFD for 14 weeks. Decreased B cell maturation in Ldlr-/-Aicda-/- mice was demonstrated by 50% reduction in splenic and aortic BAFFR expression, a key signaling component of B2 cell maturation. This was associated with increased plasma IgM in Ldlr-/-Aicda-/- compared with Ldlr-/-WT animals. Importantly, Ldlr-/-Aicda-/- mice had reduced atherosclerotic lesion area (0.20 ± 0.03mm2) compared with Ldlr-/-WT (0.30 ± 0.04mm2, P < 0.05), although no differences in plaque composition were noted between groups. In addition, immunofluorescence analysis revealed increased splenic B and T cell areas independent of cell number. AID depletion directly inhibits atherosclerotic plaque formation.


Assuntos
Aterosclerose , Citidina Desaminase , Placa Aterosclerótica , Animais , Camundongos , Aterosclerose/genética , Aterosclerose/prevenção & controle , Aterosclerose/metabolismo , Linfócitos B , Diferenciação Celular , Hidrolases/metabolismo , Imunoglobulina M/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Citidina Desaminase/genética
2.
Acta Biomater ; 143: 356-371, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35257950

RESUMO

Atherosclerosis progression is a result of chronic and non-resolving inflammation, effective treatments for which still remain to be developed. We designed and developed branched poly(ß-amino ester) nanoparticles (NPs) containing plasmid DNA encoding IL-10, a potent anti-inflammatory cytokine to atherosclerosis. The NPs (NP-VHPK) are functionalized with a targeting peptide (VHPK) specific for VCAM-1, which is overexpressed by endothelial cells at sites of atherosclerotic plaque. The anionic coating affords NP-VHPK with significantly lower toxicity than uncoated NPs in both endothelial cells and red blood cells (RBCs). Following injection of NP-VHPK in ApoE-/- mice, Cy5-labelled IL-10 significantly accumulates in both whole aortas and aortic sinus sections containing plaque compared to injection with a non-targeted control. Furthermore, IL-10 gene delivery results in an attenuation of inflammation locally at the plaque site. NP-VHPK may thus have the potential to reduce the inflammatory component of atherosclerosis in a safe and effective manner. STATEMENT OF SIGNIFICANCE: Atherosclerosis is a chronic inflammatory disease that results in the formation of lipid-laden plaques within vascular walls. Although treatments using drugs and antibodies are now beginning to address the inflammation in atherosclerosis, neither is sufficient for long-term therapy. In this paper, we introduce a strategy to deliver genes encoding the anti-inflammatory protein interleukin-10 (IL-10) in vivo. We showed that Branched Poly(ß-aminoester) carrying the IL-10 gene are able to localize specifically at the plaque via surface-functionalized targeting moieties against inflamed VCAM-1 and/or ICAM-1 and to facilitate gene transcription by ECs to increase the local concentration of the IL-10 within the plaque. To date, there is no report involving non-viral nanotechnology to provide gene-based therapies for atherosclerosis.


Assuntos
Aterosclerose , Nanopartículas , Placa Aterosclerótica , Animais , Anti-Inflamatórios/uso terapêutico , Aterosclerose/tratamento farmacológico , Aterosclerose/terapia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Inflamação/tratamento farmacológico , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Placa Aterosclerótica/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Molécula 1 de Adesão de Célula Vascular/uso terapêutico
4.
Front Cardiovasc Med ; 8: 737934, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34722670

RESUMO

Atherosclerosis is the largest contributor toward life-threatening cardiovascular events. Cellular activity and cholesterol accumulation lead to vascular remodeling and the formation of fatty plaques. Complications arise from blood clots, forming at sites of plaque development, which may detach and result in thrombotic occlusions. Vascular smooth muscle cells and macrophages play dominant roles in atherosclerosis. A firm understanding of how these cells influence and modulate each other is pivotal for a better understanding of the disease and the development of novel therapeutics. Recent studies have investigated molecular interactions between both cell types and their impact on disease progression. Here we aim to review the current knowledge. Intercellular communications through soluble factors, physical contact, and extracellular vesicles are discussed. We also present relevant background on scientific methods used to study the disease, the general pathophysiology and intracellular factors involved in phenotypic modulation of vascular smooth muscle cells. We conclude this review with a discussion of the current state, shortcomings and potential future directions of the field.

5.
Environ Health Perspect ; 129(5): 57008, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34014776

RESUMO

BACKGROUND: Epidemiologic studies indicate that early life arsenic exposures are linked to an increased risk of cardiovascular diseases. Different oxidation and methylation states of arsenic exist in the environment and are formed in vivo via the action of arsenic (+3 oxidation state) methyltransferase (As3MT). Methylated arsenicals are pro-atherogenic postnatally, but pre- and perinatal effects are unclear. This is particularly important because methylated arsenicals are known to cross the placenta. OBJECTIVES: We tested the effects of early life exposure to inorganic and methylated arsenicals on atherosclerotic plaque formation and its composition in apolipoprotein E knock-out (apoE-/-) mice and evaluated whether apoE-/- mice lacking As3MT expression were susceptible to this effect. METHODS: We exposed apoE-/- or apoE-/-/As3MT-/- mice to 200 ppb inorganic or methylated arsenic in the drinking water from conception to weaning and assessed atherosclerotic plaques in the offspring at 18 wk of age. Mixed regression models were used to estimate the mean difference in each outcome relative to controls, adjusting for sex and including a random effects term to account for within-litter clustering. RESULTS: Early life exposure to inorganic arsenic, and more profoundly methylated arsenicals, resulted in significantly larger plaques in the aortic arch and sinus in both sexes. Lipid levels in these plaques were higher without a substantial difference in macrophage numbers. Smooth muscle cell content was not altered, but collagen content was lower. Importantly, there were sex-specific differences in these observations, where males had higher lipids and lower collagen in the plaque, but females did not. In mice lacking As3MT, arsenic did not alter the plaque size, although the size was highly variable. In addition, control apoE-/-/As3MT-/- mice had significantly larger plaque size compared with control apoE-/-. CONCLUSION: This study shows that early life exposure to inorganic and methylated arsenicals is pro-atherogenic with sex-specific differences in plaque composition and a potential role for As3MT in mice. https://doi.org/10.1289/EHP8171.


Assuntos
Arsênio , Placa Aterosclerótica , Efeitos Tardios da Exposição Pré-Natal , Animais , Arsênio/toxicidade , Arsenicais , Feminino , Masculino , Metilação , Metiltransferases/genética , Metiltransferases/metabolismo , Camundongos , Camundongos Knockout , Placa Aterosclerótica/induzido quimicamente , Gravidez , Fatores Sexuais
6.
Proc Natl Acad Sci U S A ; 117(44): 27556-27565, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33077599

RESUMO

Tumor-associated macrophages (TAMs) continuously fine tune their immune modulatory properties, but how gene expression programs coordinate this immune cell plasticity is largely unknown. Selective mRNA translation, controlled by MNK1/MNK2 and mTOR pathways impinging on eIF4E, facilitates reshaping of proteomes without changes in abundance of corresponding mRNAs. Using polysome profiling developed for small samples we show that, during tumor growth, gene expression in TAMs is predominately modulated via mRNA-selective changes in translational efficiencies. These alterations in gene expression paralleled accumulation of antiinflammatory macrophages with augmented phosphorylation of eIF4E, a target of the MNK1 and MNK2 kinases, known to selectively modulate mRNA translation. Furthermore, suppression of the MNK2, but not the mTOR signaling pathway, reprogrammed antiinflammatory macrophages toward a proinflammatory phenotype with the ability to activate CD8+ T cells. Thus, selective changes of mRNA translation depending on MNK2 signaling represents a key node regulating macrophage antiinflammatory functions.


Assuntos
Macrófagos/imunologia , Neoplasias/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Células MCF-7 , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Naftiridinas/farmacologia , Neoplasias/genética , Neoplasias/patologia , Fosforilação/genética , Fosforilação/imunologia , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Evasão Tumoral/genética
7.
Immunohorizons ; 4(5): 259-273, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32434880

RESUMO

Four-and-a-half LIM domain protein 2 (FHL2) is an adaptor molecule regulating various cellular processes, including signal transduction, transcription, and cell survival. Although involved in inflammation and immune responses, its role in the germinal center reaction and B cell maturation remains unknown. We found that FHL2-/- mouse spleens displayed enlarged follicles with more B cells. When a T cell-dependent immune response was elicited using SRBC, FHL2-/- germinal center area was enhanced 2-fold compared with wild type (WT), concomitant with expanded dark zones. Nevertheless, the SRBC-induced rise in spleen IgG1 expression, and plasma IgG1 levels observed in WT were absent in FHL2-/- mice, and circulating plasma cells were also reduced in FHL2-/- This could be explained by deficient upregulation of spleen activation-induced cytidine deaminase mRNA. Interestingly, FHL2-/- B cells successfully underwent class-switch recombination in vitro, and both activation-induced cytidine deaminase induction and IgG1 response to SRBC were equivalent in B cell-deficient µMT mice transplanted with WT or FHL2-/- bone marrow, suggesting that the defects observed in FHL2-/- mice were not B cell intrinsic. However, spleen lysates from FHL2-/- mice revealed a disturbed spleen microenvironment, with reduced CXCL12 and CXCL13 levels compared with WT. Our data suggest that spleen FHL2 expression is essential for a normal germinal center reaction and proper induction of class-switch recombination in response to a T cell-dependent Ag, leading to the emergence of Ab producing plasma cells. This could be due to the regulation of spleen cytokine production by FHL2.


Assuntos
Formação de Anticorpos , Linfócitos B/imunologia , Centro Germinativo/imunologia , Imunoglobulina G/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Musculares/metabolismo , Linfócitos T/imunologia , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular , Sobrevivência Celular , Quimiocina CXCL12/metabolismo , Quimiocina CXCL13/metabolismo , Switching de Imunoglobulina , Proteínas com Homeodomínio LIM/antagonistas & inibidores , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Baço/citologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Ativação Transcricional
8.
Toxicol Sci ; 166(1): 213-218, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30376133

RESUMO

Arsenic exposure increases the risk of atherosclerosis, the gradual occlusion of the large arteries with fibro-fatty plaque. While epidemiologic data provide convincing evidence this is true at higher exposures, it is unclear whether this may occur at low arsenic exposures, near the maximum contaminant level of 10 ppb. We have previously shown that 200 ppb arsenite in the drinking water increased the atherosclerosis in apolipoprotein E knock-out (apoE-/-) mice after 13 weeks, but the effects of lower concentrations were unknown. Therefore, here, we analyzed the effects of oral exposure to arsenite from 10 to 200 ppb after 13 weeks. Importantly, we found that even at the lowest concentration of arsenite, there was a significant increase in atherosclerotic plaque size. In our previous studies, we found that arsenite exposure resulted in decreased smooth muscle cells (SMCs) and collagen within the plaque. This change is indicative of a less stable phenotype that could increase the risk of rupture and subsequently, myocardial infarct or stroke in humans. In addition, we observed that lipid increased within the plaque without concomitant increase in macrophage content, suggesting that the macrophages were retaining more lipid intracellularly. We also assessed these plaque components in apoE-/- mice exposed to 10-200 ppb arsenite. Interestingly, we observed that macrophage lipid accumulation occurred at lower concentrations than the decreased SMC/collagen content. Together these data suggest that in the apoE-/- model, low arsenite concentrations are pro-atherogenic and that macrophage lipid homeostasis is more sensitive to arsenite-induced perturbation than the SMCs.


Assuntos
Apolipoproteínas E , Arsenitos/toxicidade , Aterosclerose/induzido quimicamente , Poluentes Ambientais/toxicidade , Placa Aterosclerótica/induzido quimicamente , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Camundongos , Camundongos Knockout , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
9.
Materials (Basel) ; 11(5)2018 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-29738480

RESUMO

Nanoparticles are uniquely suited for the study and development of potential therapies against atherosclerosis by virtue of their size, fine-tunable properties, and ability to incorporate therapies and/or imaging modalities. Furthermore, nanoparticles can be specifically targeted to the atherosclerotic plaque, evading off-target effects and/or associated cytotoxicity. There has been a wealth of knowledge available concerning the use of nanotechnologies in cardiovascular disease and atherosclerosis, in particular in animal models, but with a major focus on imaging agents. In fact, roughly 60% of articles from an initial search for this review included examples of imaging applications of nanoparticles. Thus, this review focuses on experimental therapy interventions applied to and observed in animal models. Particular emphasis is placed on how nanoparticle materials and properties allow researchers to learn a great deal about atherosclerosis. The objective of this review was to provide an update for nanoparticle use in imaging and drug delivery studies and to illustrate how nanoparticles can be used for sensing and modelling, for studying fundamental biological mechanisms, and for the delivery of biotherapeutics such as proteins, peptides, nucleic acids, and even cells all with the goal of attenuating atherosclerosis. Furthermore, the various atherosclerosis processes targeted mainly for imaging studies have been summarized in the hopes of inspiring new and exciting targeted therapeutic and/or imaging strategies.

10.
Sci Rep ; 8(1): 877, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343714

RESUMO

Cells mobilized from the bone marrow can contribute to endothelial regeneration and repair. Nevertheless, cardiovascular diseases are associated with diminished numbers and function of these cells, attenuating their healing potential. Gene transfer of endothelial nitric oxide synthase (eNOS) can restore the activity of circulating cells. Furthermore, estrogen accelerates the reendothelialization capacity of early outgrowth cells (EOCs). We hypothesized that overexpressing eNOS alone or in combination with estrogen stimulation in EOCs would potentiate the beneficial effects of these cells in regulating smooth muscle cell (SMC) function. Native human EOCs did not have any effect on human coronary artery SMC (hCASMC) proliferation or migration. Transfecting EOCs with a human eNOS plasmid and/or stimulating with 17ß-estradiol (E2) increased NO production 3-fold and enhanced EOC survival. Moreover, in co-culture studies, eNOS overexpressing or E2-stimulated EOCs reduced hCASMC migration (by 23% and 56% respectively), vs. control EOCs. These effects do not implicate ERK1/2 or focal adhesion kinases. Nevertheless, NOS-EOCs had no effect on hCASMC proliferation. These results suggest that overexpressing or activating eNOS in EOCs increases their survival and enhances their capacity to regulate SMC migration through paracrine effects. These data elucidate how eNOS overexpression or activation in EOCs can prevent vascular remodeling.


Assuntos
Vasos Coronários/metabolismo , Expressão Gênica , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Comunicação Parácrina , Adulto , Apoptose , Movimento Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Células Cultivadas , Vasos Coronários/citologia , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Óxido Nítrico/biossíntese , Fosforilação , Transfecção , Adulto Jovem
11.
ACS Appl Bio Mater ; 1(3): 917-927, 2018 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-34996186

RESUMO

Poly(ß-amino esters) or PBAEs are highly efficient synthetic polymers optimized for gene delivery, a complicated process dependent on polymer properties such as hydrophobicity, charge, and degradability. The modular design of PBAEs has allowed for the identification of which polymer and nanoparticle properties significantly affect gene delivery efficiency in various cell types. However, these investigations need to be extended to more difficult-to-transfect cells such as primary endothelial cells, which hold enormous potential for atherosclerosis. Here a small library of 6 different PBAEs were screened for efficacy and safety in two types of primary endothelial cells (ECs). Nearly all polymers were more efficient than commercial transfection reagents (p < 0.05), reaching 60% and 15% transfection efficiency in human and mouse primary ECs, respectively. The top performing PBAE was used to deliver a plasmid encoding the anti-inflammatory cytokine interleukin-10 (IL-10), which has the potential to reduce inflammation in atherosclerosis. Significant increases in IL-10 mRNA and protein were detectable in ECs 72 h after transfection with PBAE:IL-10 nanoparticles. Macrophages cultured in conditioned medium from IL10-transfected ECs showed activation of anti-inflammatory signaling pathways. In addition, these macrophages secreted significantly less (25%) tumor necrosis factor α (TNFα) when challenged with lipopolysaccharide (LPS). These results underline the capabilities of PBAEs to be expanded as a fine-tunable platform for anti-inflammatory gene delivery within the context of atherosclerosis.

12.
Cardiovasc Res ; 113(14): 1753-1762, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29016715

RESUMO

AIMS: Matrix metalloproteinases (MMPs) have been implicated in the development of hypertension in animal models and humans. Mmp2 deletion did not change Ang II-induced blood pressure (BP) rise. However, whether Mmp2 knockout affects angiotensin (Ang) II-induced vascular injury has not been tested. We sought to determine whether Mmp2 knockout will prevent Ang II-induced vascular injury. METHODS AND RESULTS: A fourteen-day Ang II infusion (1000 ng/kg/min, SC) increased systolic BP, decreased vasodilatory responses to acetylcholine, induced mesenteric artery (MA) hypertrophic remodelling, and enhanced MA stiffness in wild-type (WT) mice. Ang II enhanced aortic media and perivascular reactive oxygen species generation, aortic vascular cell adhesion molecule-1 and monocyte chemotactic protein-1 expression, perivascular monocyte/macrophage and T cell infiltration, and the fraction of spleen activated CD4+CD69+ and CD8+CD69+ T cells, and Ly-6Chi monocytes. Study of intracellular signalling showed that Ang II increased phosphorylation of epidermal growth factor receptor and extracellular-signal-regulated kinase 1/2 in vascular smooth muscle cells isolated from WT mice. All these effects were reduced or prevented by Mmp2 knockout, except for systolic BP elevation. Ang II increased Mmp2 expression in immune cells infiltrating the aorta and perivascular fat. Bone marrow (BM) transplantation experiments revealed that in absence of MMP2 in immune cells, Ang II-induced BP elevation was decreased, and that when MMP2 was deficient in either immune or vascular cells, Ang II-induced endothelial dysfunction was blunted. CONCLUSIONS: Mmp2 knockout impaired Ang II-induced vascular injury but not BP elevation. BM transplantation revealed a role for immune cells in Ang II-induced BP elevation, and for both vascular and immune cell MMP2 in Ang II-induced endothelial dysfunction.


Assuntos
Angiotensina II/farmacologia , Hipertensão/genética , Metaloproteinase 2 da Matriz/genética , Lesões do Sistema Vascular/induzido quimicamente , Lesões do Sistema Vascular/genética , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/genética , Endotélio Vascular/metabolismo , Hipertensão/fisiopatologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo/genética , Lesões do Sistema Vascular/metabolismo
13.
Environ Health Perspect ; 125(7): 077001, 2017 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-28728140

RESUMO

BACKGROUND: Arsenic is metabolized through a series of oxidative methylation reactions by arsenic (3) methyltransferase (As3MT) to yield methylated intermediates. Although arsenic exposure is known to increase the risk of atherosclerosis, the contribution of arsenic methylation and As3MT remains undefined. OBJECTIVES: Our objective was to define whether methylated arsenic intermediates were proatherogenic and whether arsenic biotransformation by As3MT was required for arsenic-enhanced atherosclerosis. METHODS: We utilized the apoE−/− mouse model to compare atherosclerotic plaque size and composition after inorganic arsenic, methylated arsenical, or arsenobetaine exposure in drinking water. We also generated apoE−/−/As3mt−/− double knockout mice to test whether As3MT-mediated biotransformation was required for the proatherogenic effects of inorganic arsenite. Furthermore, As3MT expression and function were assessed in in vitro cultures of plaque-resident cells. Finally, bone marrow transplantation studies were performed to define the contribution of As3MT-mediated methylation in different cell types to the development of atherosclerosis after inorganic arsenic exposure. RESULTS: We found that methylated arsenicals, but not arsenobetaine, are proatherogenic and that As3MT is required for arsenic to induce reactive oxygen species and promote atherosclerosis. Importantly, As3MT was expressed and functional in multiple plaque-resident cell types, and transplant studies indicated that As3MT is required in extrahepatic tissues to promote atherosclerosis. CONCLUSION: Taken together, our findings indicate that As3MT acts to promote cardiovascular toxicity of arsenic and suggest that human AS3MT SNPs that correlate with enzyme function could predict those most at risk to develop atherosclerosis among the millions that are exposed to arsenic. https://doi.org/10.1289/EHP806.


Assuntos
Arsênio/toxicidade , Arsenicais/metabolismo , Aterosclerose/induzido quimicamente , Aterosclerose/genética , Expressão Gênica , Metiltransferases/genética , Poluentes Químicos da Água/toxicidade , Animais , Humanos , Masculino , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Knockout
14.
Arterioscler Thromb Vasc Biol ; 37(7): 1315-1322, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28450294

RESUMO

OBJECTIVE: Coagulation and inflammation are inter-related. Gas6 (growth arrest-specific 6) promotes venous thrombosis and participates to inflammation through endothelial-innate immune cell interactions. Innate immune cells can provide the initiating stimulus for venous thrombus development. We hypothesize that Gas6 promotes monocyte recruitment during venous thrombosis. APPROACH AND RESULTS: Deep venous thrombosis was induced in wild-type and Gas6-deficient (-/-) mice using 5% FeCl3 and flow reduction in the inferior vena cava. Total monocyte depletion was achieved by injection of clodronate before deep venous thrombosis. Inflammatory monocytes were depleted using an anti-C-C chemokine receptor type 2 (CCR2) antibody. Similarly, injection of an anti-chemokine ligand 2 (CCL2) antibody induced CCL2 depletion. Flow cytometry and immunofluorescence were used to characterize the monocytes recruited to the thrombus. In vivo, absence of Gas6 was associated with a reduction of monocyte recruitment in both deep venous thrombosis models. Global monocyte depletion by clodronate leads to smaller thrombi in wild-type mice. Compared with wild type, the thrombi from Gas6-/- mice contain less inflammatory (CCR2hiCX3CR1lo) monocytes, consistent with a Gas6-dependent recruitment of this monocyte subset. Correspondingly, selective depletion of CCR2hiCX3CR1lo monocytes reduced the formation of venous thrombi in wild-type mice demonstrating a predominant role of the inflammatory monocytes in thrombosis. In vitro, the expression of both CCR2 and CCL2 were Gas6 dependent in monocytes and endothelial cells, respectively, impacting monocyte migration. Moreover, Gas6-dependent CCL2 expression and monocyte migration were mediated via JNK (c-Jun N-terminal kinase). CONCLUSIONS: This study demonstrates that Gas6 specifically promotes the recruitment of inflammatory CCR2hiCX3CR1lo monocytes through the regulation of both CCR2 and CCL2 during deep venous thrombosis.


Assuntos
Quimiotaxia de Leucócito , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Monócitos/metabolismo , Receptores CCR2/metabolismo , Receptores de Quimiocinas/metabolismo , Veia Cava Inferior/metabolismo , Trombose Venosa/metabolismo , Animais , Receptor 1 de Quimiocina CX3C , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Ácido Clodrônico/farmacologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Predisposição Genética para Doença , Inflamação/genética , Inflamação/patologia , Inflamação/prevenção & controle , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Comunicação Parácrina , Fenótipo , Receptores CCR2/genética , Transdução de Sinais , Veia Cava Inferior/efeitos dos fármacos , Veia Cava Inferior/patologia , Trombose Venosa/genética , Trombose Venosa/patologia , Trombose Venosa/prevenção & controle
15.
Sci Rep ; 7: 43817, 2017 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-28272478

RESUMO

Erythro-myeloid progenitors (EMPs) were recently described to arise from the yolk sac endothelium, just prior to vascular remodeling, and are the source of adult/post-natal tissue resident macrophages. Questions remain, however, concerning whether EMPs differentiate directly from the endothelium or merely pass through. We provide the first evidence in vivo that EMPs can emerge directly from endothelial cells (ECs) and demonstrate a role for these cells in vascular development. We find that EMPs express most EC markers but late EMPs and EMP-derived cells do not take up acetylated low-density lipoprotein (AcLDL), as ECs do. When the endothelium is labelled with AcLDL before EMPs differentiate, EMPs and EMP-derived cells arise that are AcLDL+. If AcLDL is injected after the onset of EMP differentiation, however, the majority of EMP-derived cells are not double labelled. We find that cell division precedes entry of EMPs into circulation, and that blood flow facilitates the transition of EMPs from the endothelium into circulation in a nitric oxide-dependent manner. In gain-of-function studies, we inject the CSF1-Fc ligand in embryos and found that this increases the number of CSF1R+ cells, which localize to the venous plexus and significantly disrupt venous remodeling. This is the first study to definitively establish that EMPs arise from the endothelium in vivo and show a role for early myeloid cells in vascular development.


Assuntos
Células Endoteliais/citologia , Células Precursoras Eritroides/citologia , Células-Tronco Embrionárias Murinas/citologia , Células Progenitoras Mieloides/citologia , Remodelação Vascular , Saco Vitelino/citologia , Animais , Células Endoteliais/metabolismo , Células Precursoras Eritroides/metabolismo , Feminino , Hematopoese , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Células-Tronco Embrionárias Murinas/metabolismo , Células Progenitoras Mieloides/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Saco Vitelino/irrigação sanguínea , Saco Vitelino/embriologia
16.
Hypertension ; 67(5): 836-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27001296
17.
Thromb Haemost ; 115(3): 467-73, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26676959

RESUMO

In the developing embryo, the vasculature first takes the form of a web-like network called the vascular plexus. Arterial and venous differentiation is subsequently guided by the specific expression of genes in the endothelial cells that provide spatial and temporal cues for development. Notch1/4, Notch ligand delta-like 4 (Dll4), and Notch downstream effectors are typically expressed in arterial cells along with EphrinB2, whereas chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) and EphB4 characterise vein endothelial cells. Haemodynamic forces (blood pressure and blood flow) also contribute importantly to vascular remodelling. Early arteriovenous differentiation and local blood flow may hold the key to future inflammatory diseases. Indeed, despite the fact that atherosclerosis risk factors such as smoking, hypertension, hypercholesterolaemia, and diabetes all induce endothelial cell dysfunction throughout the vasculature, plaques develop only in arteries, and they localise essentially in vessel branch points, curvatures and bifurcations, where blood flow (and consequently shear stress) is low or oscillatory. Arterial segments exposed to high blood flow (and high laminar shear stress) tend to remain plaque-free. These observations have led many to investigate what particular properties of arterial or venous endothelial cells confer susceptibility or protection from plaque formation, and how that might interact with a particular shear stress environment.


Assuntos
Artérias/fisiologia , Aterosclerose/metabolismo , Resistência ao Cisalhamento , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Células Endoteliais/citologia , Endotélio Vascular/metabolismo , Efrina-B2/metabolismo , Humanos , Inflamação , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Fenótipo , Receptor Notch1/metabolismo , Transdução de Sinais/fisiologia , Estresse Mecânico
18.
PLoS One ; 10(9): e0136592, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26332580

RESUMO

Epidemiological studies have shown that arsenic exposure increases atherosclerosis, but the mechanisms underlying this relationship are unknown. Monocytes, macrophages and platelets play an important role in the initiation of atherosclerosis. Circulating monocytes and macrophages bind to the activated vascular endothelium and migrate into the sub-endothelium, where they become lipid-laden foam cells. This process can be facilitated by platelets, which favour monocyte recruitment to the lesion. Thus, we assessed the effects of low-to-moderate arsenic exposure on monocyte adhesion to endothelial cells, platelet activation and platelet-monocyte interactions. We observed that arsenic induces human monocyte adhesion to endothelial cells in vitro. These findings were confirmed ex vivo using a murine organ culture system at concentrations as low as 10 ppb. We found that both cell types need to be exposed to arsenic to maximize monocyte adhesion to the endothelium. This adhesion process is specific to monocyte/endothelium interactions. Hence, no effect of arsenic on platelet activation or platelet/leukocyte interaction was observed. We found that arsenic increases adhesion of mononuclear cells via increased CD29 binding to VCAM-1, an adhesion molecule found on activated endothelial cells. Similar results were observed in vivo, where arsenic-exposed mice exhibit increased VCAM-1 expression on endothelial cells and increased CD29 on circulating monocytes. Interestingly, expression of adhesion molecules and increased binding can be inhibited by antioxidants in vitro and in vivo. Together, these data suggest that arsenic might enhance atherosclerosis by increasing monocyte adhesion to endothelial cells, a process that is inhibited by antioxidants.


Assuntos
Arsênio/efeitos adversos , Aterosclerose/induzido quimicamente , Aterosclerose/patologia , Endotélio Vascular/patologia , Poluentes Ambientais/efeitos adversos , Monócitos/efeitos dos fármacos , Monócitos/patologia , Animais , Antioxidantes/farmacologia , Aterosclerose/metabolismo , Adesão Celular , Linhagem Celular , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Técnicas de Cultura de Órgãos , Ativação Plaquetária/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
19.
PLoS One ; 10(6): e0128539, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26046630

RESUMO

After Chernobyl and Fukushima Daï Chi, two major nuclear accidents, large amounts of radionuclides were released in the environment, mostly caesium 137 (137Cs). Populations living in contaminated territories are chronically exposed to radionuclides by ingestion of contaminated food. However, questions still remain regarding the effects of low dose ionizing radiation exposure on the development and progression of cardiovascular diseases. We therefore investigated the effects of a chronic internal exposure to 137Cs on atherosclerosis in predisposed ApoE-/- mice. Mice were exposed daily to 0, 4, 20 or 100 kBq/l 137Cs in drinking water, corresponding to range of concentrations found in contaminated territories, for 6 or 9 months. We evaluated plaque size and phenotype, inflammatory profile, and oxidative stress status in different experimental groups. Results did not show any differences in atherosclerosis progression between mice exposed to 137Cs and unexposed controls. However, 137Cs exposed mice developed more stable plaques with decreased macrophage content, associated with reduced aortic expression of pro-inflammatory factors (CRP, TNFα, MCP-1, IFNγ) and adhesion molecules (ICAM-1, VCAM-1 and E-selectin). Lesions of mice exposed to 137Cs were also characterized by enhanced collagen and smooth muscle cell content, concurrent with reduced matrix metalloproteinase MMP8 and MMP13 expression. These results suggest that low dose chronic exposure of 137Cs in ApoE-/- mice enhances atherosclerotic lesion stability by inhibiting pro-inflammatory cytokine and MMP production, resulting in collagen-rich plaques with greater smooth muscle cell and less macrophage content.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/patologia , Raios gama , Animais , Aorta/metabolismo , Aorta/patologia , Aorta/efeitos da radiação , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Moléculas de Adesão Celular/metabolismo , Radioisótopos de Césio/química , Colesterol/sangue , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Macrófagos/citologia , Macrófagos/imunologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo/efeitos da radiação
20.
Arterioscler Thromb Vasc Biol ; 35(6): 1472-1479, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25838425

RESUMO

OBJECTIVE: Coronary artery disease (CAD), including myocardial infarction (MI), is the main cause of death in the world. Genome-wide association studies have identified dozens of single nucleotide polymorphisms (SNPs) associated with CAD/MI. One of the most robust CAD/MI genetic associations is with intronic SNPs in the gene PHACTR1 on chromosome 6p24. How these PHACTR1 SNPs influence CAD/MI risk, and whether PHACTR1 itself is the causal gene at the locus, is currently unknown. APPROACH AND RESULTS: Using genetic fine-mapping and DNA resequencing experiments, we prioritized an intronic SNP (rs9349379) in PHACTR1 as causal variant. We showed that this variant is an expression quantitative trait locus for PHACTR1 expression in human coronary arteries. Experiments in endothelial cell extracts confirmed that alleles at rs9349379 are differentially bound by the transcription factors myocyte enhancer factor-2. We engineered a deletion of this myocyte enhancer factor-2-binding site using CRISPR/Cas9 genome-editing methodology. Heterozygous endothelial cells carrying this deletion express 35% less PHACTR1. Finally, we found no evidence that PHACTR1 expression levels are induced when stimulating human endothelial cells with vascular endothelial growth factor, tumor necrosis factor-α, or shear stress. CONCLUSIONS: Our results establish a link between intronic SNPs in PHACTR1, myocyte enhancer factor-2 binding, and transcriptional functions at the locus, PHACTR1 expression levels in coronary arteries and CAD/MI risk. Because PHACTR1 SNPs are not associated with the traditional risk factors for CAD/MI (eg, blood lipids or pressure, diabetes mellitus), our results suggest that PHACTR1 may influence CAD/MI risk through as yet unknown mechanisms in the vascular endothelium.


Assuntos
Cromossomos Humanos Par 6/genética , Vasos Coronários/metabolismo , Fatores de Transcrição MEF2/metabolismo , Proteínas dos Microfilamentos/metabolismo , Infarto do Miocárdio/genética , Polimorfismo de Nucleotídeo Único , Alelos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Infarto do Miocárdio/metabolismo , Umbigo/irrigação sanguínea , Veias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...