Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 265
Filtrar
1.
Acta sci., Health sci ; 44: e58558, Jan. 14, 2022.
Artigo em Inglês | LILACS | ID: biblio-1367771

RESUMO

Cardiovascular disease(CVD) remains the major cause of mortality in the world, typically claiming a third of all deaths. The primary cause of CVD is atherosclerosis. Therefore, timely prevention and therapy of atherosclerosis are able to reduce the risk of the development of its clinical manifestations. Anti-atherosclerotic activity of medicinal plants mainly appears in their multiple effects.This study was carried out to evaluate the hypolipidemic activity of virgin olive oil in experimentally induced hyperlipemic Wistar. A total of 24 rats were randomly allocated to 4 equal groups and treated as follows for 50 days: (1) Normal control (NC); that were fed with a standart diet; (2) High Cholesterol Diet Control (HCD); which received high cholesterol diet for 50 days; (3) Animals receiving high cholesterol diet for 50 days, after this period the animals are fed for eight days by the standard foodand receiving by gavage virgin olive oil (HCD+VOO) and(4) Animals fed for eight days with the standard food and receiving by gavage olive oil (VOO). High Cholesterol Diet containing yolk egg and coconut oil. Results showed that olive oil caused a significant (p < 0.01) reduction in serum levels of Total Cholesterol (TC), Triglycerides (TG), Low­Density Lipoprotein Cholesterol (LDL) and Atherogenic Index Serum (AIS). The results also demonstrated a significant (p < 0.01) increase in High­Density Lipoprotein Cholesterol (HDL). Moreover, virgin olive oil induced a significant reduction in liver lipid content. On the other hand, a High cholesterol diet induced oxidative stress was measured by estimating reduced glutathione level and amount of thiobarbituric acid reactive substances (TBARS) formed as an index of lipid peroxidation in a liver and a heart. Virgin olive oil supplementation attenuated all these variations. Our observations of the study indicate that the virgin olive oil has a significant antihyperlipidemic potential.


Assuntos
Animais , Ratos , Estresse Oxidativo/imunologia , Aterosclerose/dietoterapia , Dieta Hiperlipídica/métodos , Azeite de Oliva/farmacologia , Triglicerídeos/farmacologia , Peroxidação de Lipídeos/imunologia , Colesterol/farmacologia , Ratos Wistar/imunologia , Dieta Aterogênica/métodos , Glutationa/farmacologia , Hipercolesterolemia/imunologia , Lipoproteínas/imunologia
2.
Biomed Pharmacother ; 143: 112224, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34649351

RESUMO

T-cells orchestrate the inflammatory responses in atherosclerosis, and their function is modified by the lipoprotein milieu and complement activity. We investigated the effects of fluvastatin on the expression of complement decay-accelerating factor (DAF/CD55) antigen, and the levels of transcription factors in circulating T-cells in hypercholesterolemia. The hypercholesterolemic state was associated with the upregulation of DAF expression on circulating T-cells and increased levels nuclear factor kappa B (NF-kB) and interferon regulatory factor 4 (IRF4). Notably, the elevated levels of DAF and NF-kB expression persisted following treatment with fluvastatin. Therefore, the pleiotropic effects of fluvastatin are partially ascribed to its ability to mediate T-cell activation and regulate complement activity. Consequently, enhanced therapeutic interventions that targets complement-induced T-cell activation may be important in mitigating the development of atherosclerosis and major cardiovascular events in individuals with hypercholesterolemia.


Assuntos
Colesterol/sangue , Proteínas do Sistema Complemento/metabolismo , Fluvastatina/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipercolesterolemia/tratamento farmacológico , Ativação Linfocitária/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Animais , Biomarcadores/sangue , Antígenos CD55/metabolismo , Modelos Animais de Doenças , Hipercolesterolemia/imunologia , Hipercolesterolemia/metabolismo , Fatores Reguladores de Interferon/metabolismo , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 41(10): 2598-2615, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34348488

RESUMO

Objective: Hypercholesterolemia-induced NETosis and accumulation of neutrophil extracellular traps (NETs) in the atherosclerotic lesion exacerbates inflammation and is causally implicated in plaque progression. We investigated whether hypercholesterolemia additionally impairs the clearance of NETs mediated by endonucleases such as DNase1 and DNase1L3 and its implication in advanced atherosclerotic plaque progression. Approach and Results: Using a mouse model, we demonstrate that an experimental increase in the systemic level of NETs leads to a rapid increase in serum DNase activity, which is critical for the prompt clearance of NETs and achieving inflammation resolution. Importantly, hypercholesterolemic mice demonstrate an impairment in this critical NET-induced DNase response with consequent delay in the clearance of NETs and defective inflammation resolution. Administration of tauroursodeoxycholic acid, a chemical chaperone that relieves endoplasmic reticulum stress, rescued the hypercholesterolemia-induced impairment in the NET-induced DNase response suggesting a causal role for endoplasmic reticulum stress in this phenomenon. Correction of the defective DNase response with exogenous supplementation of DNase1 in Apoe-/- mice with advanced atherosclerosis resulted in a decrease in plaque NET content and significant plaque remodeling with decreased area of plaque necrosis and increased collagen content. From a translational standpoint, we demonstrate that humans with hypercholesterolemia have elevated systemic extracellular DNA levels and decreased plasma DNase activity. Conclusions: These data suggest that hypercholesterolemia impairs the NET-induced DNase response resulting in defective clearance and accumulation of NETs in the atherosclerotic plaque. Therefore, strategies aimed at rescuing this defect could be of potential therapeutic benefit in promoting inflammation resolution and atherosclerotic plaque stabilization.


Assuntos
Doenças da Aorta/etiologia , Aterosclerose/etiologia , Armadilhas Extracelulares/metabolismo , Hipercolesterolemia/complicações , Mediadores da Inflamação/metabolismo , Inflamação/etiologia , Neutrófilos/metabolismo , Placa Aterosclerótica , Animais , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Células CACO-2 , Desoxirribonuclease I/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Endodesoxirribonucleases/metabolismo , Estresse do Retículo Endoplasmático , Feminino , Células HL-60 , Células Hep G2 , Humanos , Hipercolesterolemia/imunologia , Hipercolesterolemia/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Necrose , Neutrófilos/imunologia , Transdução de Sinais , Células THP-1
4.
J Clin Lipidol ; 14(6): 850-858, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33011137

RESUMO

BACKGROUND: Accumulation of lipoprotein X (LpX) in blood can cause severe hypercholesterolemia and cutaneous xanthomas. Monocytes sensitively sense lipid changes in circulation and contribute to inflammation. However, how monocytes respond to LpX is undefined. OBJECTIVE: We examined the phenotype of monocytes from a patient, who had LpX, severe hypercholesterolemia, and extensive cutaneous xanthomas, and effects of semiselective plasmapheresis therapy (SPPT). METHOD: Fluorescence-activated cell sorting and adhesion assays were used to examine monocyte phenotype and ex vivo oxidized low-density lipoprotein uptake and adhesion in the patient before and after treatment with SPPT. Effects of plasma from the patient on the phenotype and adhesion of monocytes from a healthy participant were determined. RESULTS: SPPT improved hypercholesterolemia and cutaneous xanthomas. Before treatment, the patient had lower frequency of nonclassical monocytes but higher frequency of intermediate monocytes than the control participant. Before treatment, monocytes from the patient with LpX showed more intracellular lipid accumulation, alterations in several cell surface markers and intracellular cytokines, as well as enhanced oxidized low-density lipoprotein uptake and reduced adhesion compared with control. After SPPT, the phenotypes of monocytes from the patient with LpX were similar to control monocytes. Incubation with plasma from the patient before treatment as compared with plasma from the control participant or the patient after treatment increased CD11c expression and adhesion of monocytes from a healthy participant. CONCLUSION: LpX-induced hypercholesterolemia increased lipid accumulation and altered the phenotype of monocytes, which may contribute to cutaneous xanthoma development. Removal of LpX by SPPT reduced lipid accumulation and improved monocyte phenotype, likely contributing to xanthoma resolution.


Assuntos
Lipoproteína-X/sangue , Monócitos/citologia , Fenótipo , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/imunologia
5.
Biosci Rep ; 40(9)2020 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-32885808

RESUMO

Statins inhibit cholesterol biogenesis and modulate atheroma inflammation to reduce cardiovascular risks. Promoted by immune and non-immune cells, serum C-reactive protein (CRP) might be a biomarker suboptimal to assess inflammation status. Although it has been reported that statins modulated inflammation via microRNAs (miRNAs), evidence remains lacking on comprehensive profiling of statin-induced miRNAome alterations in immune cells. We recruited 19 hypercholesterolemic patients receiving 2 mg/day pitavastatin and 15 ones receiving 10 mg/day atorvastatin treatment for 12 weeks, and performed microarray-based profiling of 1733 human mature miRNAs in peripheral blood mononuclear cells (PBMCs) before and after statin treatment. Differentially expressed miRNAs were determined if their fold changes were >1.50 or <0.67, after validated using quantitative polymerase chain reaction (qPCR). The miRSystem and miTALOS platforms were utilized for pathway analysis. Of the 34 patients aged 63.7 ± 6.2 years, 27 were male and 19 were with coronary artery disease. We discovered that statins induced differential expressions of miR-483-5p, miR-4667-5p, miR-1244, and miR-3609, with qPCR-validated fold changes of 1.74 (95% confidence interval, 1.33-2.15), 1.61 (1.25-1.98), 1.61 (1.01-2.21), and 1.68 (1.19-2.17), respectively. The fold changes of the four miRNAs were not correlated with changes of low-density-lipoprotein cholesterol or CRP, after sex, age, and statin type were adjusted. We also revealed that RhoA and transforming growth factor-ß signaling pathways might be regulated by the four miRNAs. Given our findings, miRNAs might be involved in statin-induced inflammation modulation in PBMCs, providing likelihood to assess and reduce inflammation in patients with atherosclerotic cardiovascular diseases.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipercolesterolemia/tratamento farmacológico , MicroRNAs/metabolismo , Placa Aterosclerótica/tratamento farmacológico , Idoso , Atorvastatina/farmacologia , Atorvastatina/uso terapêutico , LDL-Colesterol/sangue , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipercolesterolemia/sangue , Hipercolesterolemia/complicações , Hipercolesterolemia/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Placa Aterosclerótica/sangue , Placa Aterosclerótica/imunologia , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Taiwan
6.
Int Rev Neurobiol ; 154: 325-344, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32739010

RESUMO

Alzheimer's disease is characterized by aggregated amyloid beta plaques and neurofibrillary tangles. Apart from the plaques and tangles, microglial activation plays a significant role in neurodegeneration and neuronal function. This review discusses the way in which microglial activation influences neurodegeneration and how systemic inflammation, type 2 diabetes mellitus, obesity and hypercholesterolemia influence neuroinflammation. Also reviewed is how systemic inflammation influences microglial activation along with the relationship between microglial activation and glucose metabolism.


Assuntos
Citocinas , Diabetes Mellitus Tipo 2 , Glucose/metabolismo , Hipercolesterolemia , Inflamação , Síndrome Metabólica , Microglia , Doenças Neurodegenerativas , Obesidade , Animais , Citocinas/imunologia , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipercolesterolemia/imunologia , Hipercolesterolemia/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Síndrome Metabólica/imunologia , Síndrome Metabólica/metabolismo , Microglia/imunologia , Microglia/metabolismo , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/metabolismo , Obesidade/imunologia , Obesidade/metabolismo
7.
Clin Immunol ; 215: 108450, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32360516

RESUMO

Acute respiratory distress syndrome (ARDS) is a devastating clinical manifestation of COVID-19 pneumonia and is mainly based on an immune-driven pathology. Mounting evidence suggests that COVID-19 is fueled by a maladaptive host inflammatory response that involves excessive activation of innate immune pathways. While a "cytokine storm" involving IL-6 and other cytokines has been documented, complement C3 activation has been implicated as an initial effector mechanism that exacerbates lung injury in preclinical models of SARS-CoV infection. C3-targeted intervention may provide broader therapeutic control of complement-mediated inflammatory damage in COVID-19 patients. Herein, we report the clinical course of a patient with severe ARDS due to COVID-19 pneumonia who was safely and successfully treated with the compstatin-based complement C3 inhibitor AMY-101.


Assuntos
Betacoronavirus/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Complemento C3/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Infecções por Coronavirus/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Pneumonia Viral/tratamento farmacológico , Idoso , Antivirais/uso terapêutico , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/imunologia , Fibrilação Atrial/patologia , Fibrilação Atrial/virologia , Betacoronavirus/imunologia , Betacoronavirus/patogenicidade , COVID-19 , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Humanos , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/imunologia , Hipercolesterolemia/patologia , Hipercolesterolemia/virologia , Hipertensão/tratamento farmacológico , Hipertensão/imunologia , Hipertensão/patologia , Hipertensão/virologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Pandemias , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Pneumonia Viral/virologia , SARS-CoV-2 , Resultado do Tratamento
8.
Mol Nutr Food Res ; 64(15): e1900732, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31389129

RESUMO

SCOPE: Akkermansia muciniphila (A. muciniphila) is an intestinal commensal with anti-inflammatory properties both in the intestine and other organs. The aim is to investigate the effects of oral administration of A. muciniphila on lipid metabolism, immunity, and cuff-induced neointima formation in hyperlipidemic APOE*3-Leiden (E3L).CETP mice. METHODS AND RESULTS: Hyperlipidemic male E3L.CETP mice are daily treated with 2 × 108 CFU A. muciniphila by oral gavage for 4 weeks and the effects are determined on plasma lipid levels, immune parameters, and cuff-induced neointima formation and composition. A. muciniphila administration lowers body weight and plasma total cholesterol and triglycerides levels. A. muciniphila influences the immune cell composition in mesenteric lymph nodes, as evident from an increased total B cell population, while reducing the total T cell and neutrophil populations. Importantly, A. muciniphila reduces the expression of the activation markers MHCII on dendritic cells and CD86 on B cells. A. muciniphila also increases whole blood ex vivo lipopolysaccharide-stimulated IL-10 release. Finally, although treatment with A. muciniphila improves lipid metabolism and immunity, it does not affect neointima formation or composition. CONCLUSIONS: Four weeks of treatment with A. muciniphila exerts lipid-lowering and immunomodulatory effects, which are insufficient to inhibit neointima formation in hyperlipidemic E3L.CETP mice.


Assuntos
Hiperlipidemias/terapia , Fatores Imunológicos/farmacologia , Lipídeos/sangue , Probióticos/administração & dosagem , Administração Oral , Akkermansia/imunologia , Akkermansia/fisiologia , Animais , Apolipoproteína E3/genética , Modelos Animais de Doenças , Hipercolesterolemia/imunologia , Hipercolesterolemia/metabolismo , Hipercolesterolemia/terapia , Hiperlipidemias/imunologia , Hiperlipidemias/metabolismo , Metabolismo dos Lipídeos , Lipopolissacarídeos/sangue , Linfonodos/imunologia , Masculino , Camundongos Mutantes , Neointima/etiologia , Neointima/prevenção & controle
9.
Nat Immunol ; 21(1): 42-53, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31768073

RESUMO

Pathogen-associated molecular patterns (PAMPs) have the capacity to couple inflammatory gene expression to changes in macrophage metabolism, both of which influence subsequent inflammatory activities. Similar to their microbial counterparts, several self-encoded damage-associated molecular patterns (DAMPs) induce inflammatory gene expression. However, whether this symmetry in host responses between PAMPs and DAMPs extends to metabolic shifts is unclear. Here, we report that the self-encoded oxidized phospholipid oxPAPC alters the metabolism of macrophages exposed to lipopolysaccharide. While cells activated by lipopolysaccharide rely exclusively on glycolysis, macrophages exposed to oxPAPC also use mitochondrial respiration, feed the Krebs cycle with glutamine, and favor the accumulation of oxaloacetate in the cytoplasm. This metabolite potentiates interleukin-1ß production, resulting in hyperinflammation. Similar metabolic adaptions occur in vivo in hypercholesterolemic mice and human subjects. Drugs that interfere with oxPAPC-driven metabolic changes reduce atherosclerotic plaque formation in mice, thereby underscoring the importance of DAMP-mediated activities in pathophysiological conditions.


Assuntos
Alarminas/imunologia , Lipopolissacarídeos/imunologia , Macrófagos/metabolismo , Moléculas com Motivos Associados a Patógenos/imunologia , Fosfatidilcolinas/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Glicólise/fisiologia , Hipercolesterolemia/imunologia , Hipercolesterolemia/patologia , Inflamação/prevenção & controle , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Fosforilação Oxidativa , Placa Aterosclerótica/patologia , Placa Aterosclerótica/prevenção & controle
10.
Cardiovasc Res ; 115(3): 510-518, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30629143

RESUMO

Since the discovery of the role of proprotein convertase subtilisin kexin 9 (PCSK9) in the regulation of low-density lipoprotein cholesterol (LDL-C) in 2003, a paradigm shift in the treatment of hypercholesterolaemia has occurred. The PCSK9 secreted into the circulation is a major downregulator of the low-density lipoprotein receptor (LDLR) protein, as it chaperones it to endosomes/lysosomes for degradation. Humans with loss-of-function of PCSK9 exhibit exceedingly low levels of LDL-C and are protected from atherosclerosis. As a consequence, innovative strategies to modulate the levels of PCSK9 have been developed. Since 2015 inhibitory monoclonal antibodies (evolocumab and alirocumab) are commercially available. When subcutaneously injected every 2-4 weeks, they trigger a ∼60% LDL-C lowering and a 15% reduction in the risk of cardiovascular events. Another promising approach consists of a liver-targetable specific PCSK9 siRNA which results in ∼50-60% LDL-C lowering that lasts up to 6 months (Phases II-III clinical trials). Other strategies under consideration include: (i) antibodies targeting the C-terminal domain of PCSK9, thereby inhibiting the trafficking of PCSK9-LDLR to lysosomes; (ii) small molecules that either prevent PCSK9 binding to the LDLR, its trafficking to lysosomes or its secretion from cells; (iii) complete silencing of PCSK9 by CRISPR-Cas9 strategies; (iv) PCSK9 vaccines that inhibit the activity of circulating PCSK9. Time will tell whether other strategies can be as potent and safe as monoclonal antibodies to lower LDL-C levels.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticolesterolemiantes/uso terapêutico , Colesterol/sangue , Terapia Genética/métodos , Hipercolesterolemia/terapia , Pró-Proteína Convertase 9 , Inibidores de Serina Proteinase/uso terapêutico , Vacinas/uso terapêutico , Animais , Anticorpos Monoclonais/efeitos adversos , Anticolesterolemiantes/efeitos adversos , Biomarcadores/sangue , Inativação Gênica , Marcação de Genes , Terapia Genética/efeitos adversos , Humanos , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Hipercolesterolemia/imunologia , Inibidores de PCSK9 , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/imunologia , Pró-Proteína Convertase 9/metabolismo , Receptores de LDL/antagonistas & inibidores , Receptores de LDL/metabolismo , Inibidores de Serina Proteinase/efeitos adversos , Resultado do Tratamento , Vacinas/efeitos adversos
11.
J Immunol ; 202(5): 1531-1539, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30683705

RESUMO

Mast cells (MCs) are potent innate immune cells that aggravate atherosclerosis through the release of proinflammatory mediators inside atherosclerotic plaques. Similarly, CD4+ T cells are constituents of the adaptive immune response and accumulate within the plaques following lipid-specific activation by APCs. Recently it has been proposed that these two cell types can interact in a direct manner. However, no indication of such an interaction has been investigated in the context of atherosclerosis. In our study, we aimed to examine whether MCs can act as APCs in atherosclerosis, thereby modulating CD4+ T cell responses. We observed that MCs increased their MHC class II expression under hyperlipidemic conditions both in vivo and in vitro. Furthermore, we showed that MCs can present Ags in vivo via MHC class II molecules. Serum from high-fat diet-fed mice also enhanced the expression of the costimulatory molecule CD86 on cultured MCs, whereas OVA peptide-loaded MCs increased OT-II CD4+ T cell proliferation in vitro. The aortic CD4+ and TH1 cell content of atherosclerotic mice that lack MCs was reduced as compared with their wild-type counterparts. Importantly, we identified MCs that express HLA-DR in advanced human atheromata, indicating that these cells are capable of Ag presentation within human atherosclerotic plaques. Therefore, in this artice, we show that MCs may directly modulate adaptive immunity by acting as APCs in atherosclerosis.


Assuntos
Aterosclerose/imunologia , Linfócitos T CD4-Positivos/imunologia , Hipercolesterolemia/imunologia , Mastócitos/imunologia , Animais , Células Cultivadas , Humanos , Masculino , Camundongos , Camundongos Knockout
12.
Med Sci (Paris) ; 35(12): 1014-1016, 2019 Dec.
Artigo em Francês | MEDLINE | ID: mdl-31903910

RESUMO

The use of monoclonal antibodies in cardiovascular diseases and metabolic disorders is still in its infancy. Recent development of anti-PCSK9 monoclonal antibodies for the treatment of dyslipidemia and of patients in secondary prevention is a breakthrough in the field. Anti- PCSK9 antibodies significantly improved LDL cholesterol reduction in patients with familial hypercholesterolemia. These antibodies have also demonstrated a significant reduction of clinical events in patients with previously established atherosclerotic disease such as myocardial infarction, ischemia stroke or peripheral artery disease. Other targets are under investigation such as inflammatory cells and cytokines to reduce atherosclerosis or myocardial lesions following myocardial infarction.


TITLE: Place des anticorps thérapeutiques dans les maladies cardiovasculaires et métaboliques aujourd'hui. ABSTRACT: La place des anticorps thérapeutiques dans les maladies cardiovasculaires et métaboliques est encore modeste en 2019 en comparaison à leur incroyable développement dans d'autres champs pathologiques. Cependant, l'arrivée récente des anticorps anti-PCSK9 (proprotein convertase subtilisin/kexin de type 9) dans l'arsenal thérapeutique va probablement changer la donne. Ces anticorps permettent non seulement d'améliorer la prise en charge des patients porteurs d'hypercholestérolémie familiale mais également de réduire le risque de complications cliniques de l'athérosclérose en prévention secondaire après un infarctus du myocarde, une artériopathie périphérique ou un accident vasculaire cérébral ischémique. D'autres stratégies thérapeutiques sont en cours d'investigation, ciblant notamment des cellules et cytokines impliquées dans les réponses immuno-inflammatoires avec pour objectif de prévenir les complications de l'athérosclérose ou les lésions du myocarde au décours d'un infarctus.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Metabólicas/tratamento farmacológico , Doenças Cardiovasculares/imunologia , Dislipidemias/tratamento farmacológico , Dislipidemias/imunologia , Humanos , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/imunologia , Doenças Metabólicas/imunologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/imunologia , Pró-Proteína Convertase 9/imunologia
13.
Iran J Allergy Asthma Immunol ; 18(5): 554-566, 2019 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-32245299

RESUMO

Many observations showed that hypercholesterolemia can disrupt immune response. Statin drugs that were used for the treatment of hypercholesterolemia patients can interfere in the regulation of the immune response and cytokine secretion. The primary aim of the current study was to investigate the immune response among treatment-naïve patients with hypercholesterolemia and healthy subjects. The secondary goal of the study was to determine whether atorvastatin can reverse the detrimental effect of hypercholesterolemia on the immune system. Peripheral blood mononuclear cells (PBMCs) were isolated from 50 patients afflicted with hypercholesterolemia who were treatment-naïve along with 50 sex/age-matched hypercholesterolemia patients receiving atorvastatin, and 50 sex/age-matched healthy subjects. Quantitative PCR and ELISA methods were used for gene and protein expression analysis of T helper 1 (Th1) and Th2 related cytokines. Additionally, the expression of the cluster of differentiation (CD) markers on T, B, and natural killer (NK) cells was measured by flow cytometry method. The results showed that hypercholesterolemia and atorvastatin down-regulated the expression of Th1-related cytokines and elevated the levels of Th2-related cytokines. The expression of cell surface markers, CD25 and CD69, was significantly decreased in the treatment-naïve, and atorvastatin groups. It seems that atorvastatin is not able to repair the deleterious effects of hypercholesterolemia on the immune system. Moreover, elevated levels of cholesterol along with the administration of atorvastatin tilt the Th1/Th2 balance in favor of Th2 and reduce T cell activation.


Assuntos
Atorvastatina/imunologia , Hipercolesterolemia/imunologia , Fatores Imunológicos/imunologia , Adulto , Antígenos CD/imunologia , Colesterol/imunologia , Citocinas/imunologia , Feminino , Humanos , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/imunologia , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade , Células Th1/imunologia , Células Th2/imunologia
14.
Sci Rep ; 8(1): 16515, 2018 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-30409998

RESUMO

Gut microbiota have been implicated in the development of atherosclerosis and cardiovascular disease. Since the prebiotic inulin is thought to beneficially affect gut microbiota, we aimed to determine the effect of inulin supplementation on atherosclerosis development in APOE*3-Leiden.CETP (E3L.CETP) mice. Female E3L.CETP mice were fed a western-type diet containing 0.1% or 0.5% cholesterol with or without 10% inulin. The effects of inulin were determined on: microbiota composition, cecal short-chain fatty acid (SCFA) levels, plasma lipid levels, atherosclerosis development, hepatic morphology and hepatic inflammation. Inulin with 0.5% dietary cholesterol increased specific bacterial genera and elevated levels of cecal SCFAs, but did not affect plasma cholesterol levels or atherosclerosis development. Surprisingly, inulin resulted in mild hepatic inflammation as shown by increased expression of inflammation markers. However, these effects were not accompanied by increased hepatic macrophage number. Analogously, inulin induced mild steatosis and increased hepatocyte size, but did not affect hepatic triglyceride content. Inulin with 0.1% dietary cholesterol did not affect hepatic morphology, nor hepatic expression of inflammation markers. Overall, inulin did not reduce hypercholesterolemia or atherosclerosis development in E3L.CETP mice despite showing clear prebiotic activity, but resulted in manifestations of hepatic inflammation when combined with a high percentage of dietary cholesterol.


Assuntos
Apolipoproteína E3/genética , Aterosclerose/imunologia , Bactérias/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Hipercolesterolemia/imunologia , Inulina/administração & dosagem , Prebióticos/administração & dosagem , Animais , Apolipoproteína E3/metabolismo , Aterosclerose/genética , Aterosclerose/metabolismo , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , DNA Bacteriano/genética , DNA Ribossômico/genética , Dieta Ocidental/efeitos adversos , Modelos Animais de Doenças , Ácidos Graxos/química , Feminino , Hipercolesterolemia/induzido quimicamente , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Inulina/farmacologia , Lipídeos/sangue , Camundongos , Camundongos Transgênicos , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
15.
J Clin Invest ; 128(6): 2370-2375, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29708512

RESUMO

Emerging data suggest that hypercholesterolemia has stimulatory effects on adaptive immunity and that these effects can promote atherosclerosis and perhaps other inflammatory diseases. However, research in this area has relied primarily on inbred strains of mice whose adaptive immune system can differ substantially from that of humans. Moreover, the genetically induced hypercholesterolemia in these models typically results in plasma cholesterol levels that are much higher than those in most humans. To overcome these obstacles, we studied human immune system-reconstituted mice (hu-mice) rendered hypercholesterolemic by treatment with adeno-associated virus 8-proprotein convertase subtilisin/kexin type 9 (AAV8-PCSK9) and a high-fat/high-cholesterol Western-type diet (WD). These mice had a high percentage of human T cells and moderate hypercholesterolemia. Compared with hu-mice that had lower plasma cholesterol, the PCSK9-WD mice developed a T cell-mediated inflammatory response in the lung and liver. Human CD4+ and CD8+ T cells bearing an effector memory phenotype were significantly elevated in the blood, spleen, and lungs of PCSK9-WD hu-mice, whereas splenic and circulating regulatory T cells were reduced. These data show that moderately high plasma cholesterol can disrupt human T cell homeostasis in vivo. This process may not only exacerbate atherosclerosis, but also contribute to T cell-mediated inflammatory diseases in the hypercholesterolemia setting.


Assuntos
Aterosclerose/imunologia , Linfócitos T CD8-Positivos/imunologia , Hipercolesterolemia/imunologia , Pró-Proteína Convertase 9/imunologia , Linfócitos T Reguladores/imunologia , Animais , Aterosclerose/patologia , Linfócitos T CD8-Positivos/patologia , Dependovirus , Humanos , Hipercolesterolemia/patologia , Camundongos , Linfócitos T Reguladores/patologia
16.
J Cardiovasc Pharmacol Ther ; 23(6): 509-517, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29764192

RESUMO

Statins are mainstream therapy in the treatment and prevention of cardiovascular disease through inhibitory effects on cholesterol synthesis. However, statins' beneficial effects in cardiovascular disease may also be attributable to their role as anti-inflammatory mediators. Here, we investigated the effects of simvastatin treatment on expression levels of interleukin (IL) 1ß in both patient with hyperlipidemia and healthy human peripheral blood mononuclear cells (PBMCs) using cholesterol crystals (CC), a cardiovascular pathogenic stimulus for activation of the NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome. Cholesterol crystal-induced NLRP3 inflammasome activation was used to trigger maturation and release of IL-1ß in PBMCs. Specifically, isolated PBMCs from patients with hyperlipidemia at baseline and following 8 weeks of in vivo treatment with simvastatin (10-20 mg) daily were stimulated with lipopolysaccharide (LPS; 100 ng/mL) for 3 hours to induce proIL-Iß expression followed by CC (2 mg/mL) stimulation for further 18 hours to activate the NLRP3 inflammasome complex to induce maturation/activation of IL-1ß. Peripheral blood mononuclear cells were also isolated from healthy donors and stimulated in vitro with simvastatin (50, 25, 5, and 2 µmol/L) prior to stimulation with LPS and CC as described above. The effects of simvastatin treatment on levels of IL-1ß expression were determined by enzyme-linked immunosorbent assay and western blot. Both in vitro and in vivo treatments with simvastatin led to a significant reduction in the levels of expression of IL-1ß in response to stimulation with CC. Simvastatin inhibits the expression and activation of IL-1ß induced by CC in PBMCs, which may contribute to its protective role in patients with cardiovascular disease.


Assuntos
Anti-Inflamatórios/uso terapêutico , Colesterol/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Hipercolesterolemia/tratamento farmacológico , Interleucina-1beta/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Sinvastatina/uso terapêutico , Adulto , Idoso , Células Cultivadas , Cristalização , Feminino , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/imunologia , Inflamassomos/agonistas , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interleucina-1beta/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína 3 que Contém Domínio de Pirina da Família NLR/agonistas , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Via Secretória/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
17.
Int J Mol Med ; 41(4): 1799-1808, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29393350

RESUMO

Hypercholesterolemia is one of primary risk factors of cardiovascular disease, together with metabolic syndrome, hypertension and diabetes. Although progress has been made, the search for novel methods of preventing and treating dyslipidemia is ongoing and current therapies for cardiovascular disease induce various side effects. ß­glucans are linear unbranched polysaccharides found in various natural sources, such as mushrooms. Due to their structure they are able to interact with innate immunity receptors, however they also act as dietary fibers in the digestive tract. As there are two forms of ß­glucans, insoluble and soluble forms, they are able to interact with lipids and biliary salts in the bowel and consequently reduce cholesterol levels. Therefore, they may be developed as a suitable therapeutic option to treat patients with dyslipidemia, as they are natural molecules that do not induce any significant side effects. The current review discusses the evidence supporting the effects of ß­glucans on cholesterol levels.


Assuntos
Anticolesterolemiantes/uso terapêutico , Colesterol/metabolismo , Fibras na Dieta/uso terapêutico , Hipercolesterolemia/metabolismo , Hipercolesterolemia/terapia , Fatores Imunológicos/uso terapêutico , beta-Glucanas/uso terapêutico , Animais , Anticolesterolemiantes/química , Colesterol/sangue , Colesterol/imunologia , Fibras na Dieta/análise , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/imunologia , Fatores Imunológicos/química , beta-Glucanas/química
18.
J Cell Mol Med ; 22(3): 1366-1382, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29364567

RESUMO

Monocytosis and neutrophilia are frequent events in atherosclerosis. These phenomena arise from the increased proliferation of hematopoietic stem and multipotential progenitor cells (HSPCs) and HSPC mobilization from the bone marrow to other immune organs and circulation. High cholesterol and inflammatory signals promote HSPC proliferation and preferential differentiation to the myeloid precursors (i.e., myelopoiesis) that than give rise to pro-inflammatory immune cells. These cells accumulate in the plaques thereby enhancing vascular inflammation and contributing to further lesion progression. Studies in animal models of atherosclerosis showed that manipulation with HSPC proliferation and differentiation through the activation of LXR-dependent mechanisms and restoration of cholesterol efflux may have a significant therapeutic potential.


Assuntos
Aterosclerose/imunologia , Colesterol/imunologia , Hipercolesterolemia/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Placa Aterosclerótica/imunologia , Animais , Aterosclerose/genética , Aterosclerose/patologia , Medula Óssea/imunologia , Medula Óssea/patologia , Diferenciação Celular , Proliferação de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Receptores X do Fígado/genética , Receptores X do Fígado/imunologia , Camundongos , Monócitos/patologia , Células-Tronco Multipotentes/imunologia , Células-Tronco Multipotentes/patologia , Neutrófilos/patologia , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/deficiência , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/imunologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia
19.
Sci Rep ; 7(1): 15655, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29142309

RESUMO

Hypercholesterolemia promotes the inflammation against lipoproteins in atherosclerosis. Development of atherosclerosis is affected by the balance between pro-inflammatory effector T cells and anti-inflammatory regulatory T (Treg) cells. However, phenotype and function of T cell subpopulations in hypercholesterolemia remain to be investigated. Here, we found that cholesterol-containing diet increased the expression of the Treg cell lineage-defining transcription factor FoxP3 among thymocytes and splenocytes. Hypercholesterolemia elevated the FoxP3 expression level and population size of peripheral Treg cells, but did not prevent enhanced proliferation of stimulated T cells. Moreover, cholesterol supplementation in diet as well as in cell culture medium promoted T cell antigen receptor (TCR) signaling in CD4+ T cells. Our results demonstrate that hypercholesterolemia enhances TCR stimulation, Treg cell development as well as T cell proliferation. Thus, our findings may help to understand why hypercholesterolemia correlates with altered CD4+ T cell responses.


Assuntos
Aterosclerose/imunologia , Fatores de Transcrição Forkhead/genética , Hipercolesterolemia/imunologia , Inflamação/imunologia , Receptores de Antígenos de Linfócitos T/genética , Animais , Aterosclerose/genética , Aterosclerose/patologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linhagem da Célula/imunologia , Colesterol na Dieta/farmacologia , Fatores de Transcrição Forkhead/imunologia , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Inflamação/genética , Inflamação/patologia , Lipoproteínas/metabolismo , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de LDL/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Timócitos/metabolismo , Timócitos/patologia
20.
Sci Rep ; 7(1): 12534, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28970592

RESUMO

Vaccination provides a promising approach for treatment of hypercholesterolemia and improvement in compliance. In this study, the appropriate virus-like particle (VLP)-peptide vaccines targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) were screened. The screening criteria of target peptides were as follows: (1) located in catalytic domain of PCSK9, or regulating the binding of PCSK9 and LDL receptors (LDLR); (2) having low/no-similarity when matched with the host proteome; (3) possessing ideal antigenicity and hydrophilicity; (4) including the functional mutation site of PCSK9. It was found that mice vaccinated with VLP -PCSK9 peptide vaccines, especially PCSK9Qß-003 vaccine, developed high titer IgG antibodies against PCSK9. PCSK9Qß-003 vaccine obviously decreased plasma total cholesterol in both Balb/c mice and LDLR+/- mice. Also, PCSK9Qß-003 vaccine decreased plasma PCSK9 level and up-regulated LDLR expression in liver. Additionally, PCSK9Qß-003 vaccine injection was associated with significant up-regulation of sterol-regulatory element-binding protein-2 (SREBP-2), hepatocyte nuclear factor 1α (HNF-1α), and 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase in LDLR+/- mice. No obvious immune injury was detected in vaccinated animals. The PCSK9Qß-003 vaccine, therefore, may be an attractive treatment approach for hypercholesterolemia through decreasing cholesterol and regulating lipid homeostasis.


Assuntos
Hipercolesterolemia/tratamento farmacológico , Pró-Proteína Convertase 9/genética , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Domínio Catalítico/efeitos dos fármacos , Colesterol/sangue , Modelos Animais de Doenças , Regulação da Expressão Gênica , Fator 1-alfa Nuclear de Hepatócito/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Hipercolesterolemia/genética , Hipercolesterolemia/imunologia , Hipercolesterolemia/prevenção & controle , Imunogenicidade da Vacina/genética , Imunogenicidade da Vacina/imunologia , Imunoglobulina G/sangue , Camundongos , Camundongos Knockout , Mutação , Inibidores de PCSK9 , Fosfoproteínas Fosfatases/genética , Receptores de LDL/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...