Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Nanobiotechnology ; 22(1): 223, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702815

RESUMO

Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.


Assuntos
Dependovirus , Vetores Genéticos , Miocárdio , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Dependovirus/genética , Animais , Nanopartículas/química , Camundongos , Miocárdio/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Humanos , Camundongos Endogâmicos C57BL , Coração , Terapia Genética/métodos , Técnicas de Transferência de Genes , Fígado/metabolismo , Tropismo Viral , Células HEK293
2.
Nano Lett ; 23(21): 9690-9696, 2023 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-37884274

RESUMO

Antiphospholipid syndrome (APS) is an autoimmune disease characterized by recurrent arterial, venous, and microvascular thrombosis where activated neutrophils play a determinant role. However, neutrophils are challenging to target given their short lifespan in circulation and spontaneous activation. Screening of a small library of gold nanoparticles (AuNPs) led to the discovery of a formulation capable of targeting activated neutrophil attachment and has demonstrated that star-shaped, anti-PSGL-1-antibody-coated AuNPs (aPSGL-1-AuNPs) were more efficacious compared with other shapes of AuNPs. Our findings further revealed an exciting and safe targeting mode toward activated neutrophils in the APS mouse model induced by human-patient-derived antiphospholipid IgGs. Our studies demonstrate that targeting is dependent on the specific topographical features of the highly segregated PSGL-1 on the activated neutrophil surface for which a high affinity shape-driven nanomedicine can be designed and implemented. As such, star-shaped aPSGL-1-AuNPs serve as a promising nanoimmunotherapy for immunothrombosis associated with neutrophil adhesion in APS.


Assuntos
Síndrome Antifosfolipídica , Nanopartículas Metálicas , Trombose , Animais , Camundongos , Humanos , Síndrome Antifosfolipídica/tratamento farmacológico , Síndrome Antifosfolipídica/complicações , Síndrome Antifosfolipídica/diagnóstico , Neutrófilos , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Trombose/tratamento farmacológico , Imunoglobulina G
3.
J Immunol ; 197(6): 2390-9, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27534554

RESUMO

During Gram-negative bacterial infections, excessive LPS induces inflammation and sepsis via action on immune cells. However, the bulk of LPS can be cleared from circulation by the liver. Liver clearance is thought to be a slow process mediated exclusively by phagocytic resident macrophages, Kupffer cells (KC). However, we discovered that LPS disappears rapidly from the circulation, with a half-life of 2-4 min in mice, and liver eliminates about three quarters of LPS from blood circulation. Using microscopic techniques, we found that ∼75% of fluor-tagged LPS in liver became associated with liver sinusoidal endothelial cells (LSEC) and only ∼25% with KC. Notably, the ratio of LSEC-KC-associated LPS remained unchanged 45 min after infusion, indicating that LSEC independently processes the LPS. Most interestingly, results of kinetic analysis of LPS bioactivity, using modified limulus amebocyte lysate assay, suggest that recombinant factor C, an LPS binding protein, competitively inhibits high-density lipoprotein (HDL)-mediated LPS association with LSEC early in the process. Supporting the previous notion, 3 min postinfusion, 75% of infused fluorescently tagged LPS-HDL complex associates with LSEC, suggesting that HDL facilitates LPS clearance. These results lead us to propose a new paradigm of LSEC and HDL in clearing LPS with a potential to avoid inflammation during sepsis.


Assuntos
Células Endoteliais/fisiologia , Lipopolissacarídeos/sangue , Lipopolissacarídeos/metabolismo , Lipoproteínas HDL/metabolismo , Fígado/citologia , Proteínas de Fase Aguda/imunologia , Proteínas de Fase Aguda/metabolismo , Animais , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Células Endoteliais/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Meia-Vida , Inflamação/imunologia , Inflamação/prevenção & controle , Cinética , Células de Kupffer/imunologia , Lipopolissacarídeos/imunologia , Lipoproteínas HDL/imunologia , Fígado/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Sepse/imunologia
4.
Circ Res ; 116(8): 1491-504, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25858071

RESUMO

The discovery of incretin-based medications represents a major therapeutic advance in the pharmacological management of type 2 diabetes mellitus (T2DM), as these agents avoid hypoglycemia, weight gain, and simplify the management of T2DM. Dipeptidyl peptidase-4 (CD26, DPP4) inhibitors are the most widely used incretin-based therapy for the treatment of T2DM globally. DPP4 inhibitors are modestly effective in reducing HbA1c (glycated hemoglobin) (≈0.5%) and while these agents were synthesized with the understanding of the role that DPP4 plays in prolonging the half-life of incretins such as glucagon-like peptide-1 and gastric inhibitory peptide, it is now recognized that incretins are only one of many targets of DPP4. The widespread expression of DPP4 on blood vessels, myocardium, and myeloid cells and the nonenzymatic function of CD26 as a signaling and binding protein, across a wide range of species, suggest a teleological role in cardiovascular regulation and inflammation. Indeed, DPP4 is upregulated in proinflammatory states including obesity, T2DM, and atherosclerosis. Consistent with this maladaptive role, the effects of DPP4 inhibition seem to exert a protective role in cardiovascular disease at least in preclinical animal models. Although 2 large clinical trials suggest a neutral effect on cardiovascular end points, current limitations of performing trials in T2DM over a limited time horizon on top of maximal medical therapy must be acknowledged before rendering judgment on the cardiovascular efficacy of these agents. This review will critically review the science of DPP4 and the effects of DPP4 inhibitors on the cardiovascular system.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Inflamação/tratamento farmacológico , Animais , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/enzimologia , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/enzimologia , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Avaliação Pré-Clínica de Medicamentos , Humanos , Inflamação/diagnóstico , Inflamação/enzimologia , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
5.
Circ Res ; 115(9): 770-780, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25186795

RESUMO

RATIONALE: Air pollution exposure has been shown to potentiate plaque progression in humans and animals. Our previous studies have suggested a role for oxidized lipids in mediating adverse vascular effect of air pollution. However, the types of oxidized lipids formed in response to air pollutants and how this occurs and their relevance to atherosclerosis are not fully understood. OBJECTIVE: To investigate the mechanisms by which particulate matter <2.5 µm (PM2.5) induces progression of atherosclerosis. METHODS AND RESULTS: Atherosclerosis-prone ApoE(-/-) or LDLR(-/-) mice were exposed to filtered air or concentrated ambient PM2.5 using a versatile aerosol concentrator enrichment system for 6 months. PM2.5 increased 7-ketocholesterol (7-KCh), an oxidatively modified form of cholesterol, in plasma intermediate density lipoprotein/low-density lipoprotein fraction and in aortic plaque concomitant with progression of atherosclerosis and increased CD36 expression in plaque macrophages from PM2.5-exposed mice. Macrophages isolated from PM2.5-exposed mice displayed increased uptake of oxidized lipids without alterations in their efflux capacity. Consistent with these finding, CD36-positive macrophages displayed a heightened capacity for oxidized lipid uptake. Deficiency of CD36 on hematopoietic cells diminished the effect of air pollution on 7-KCh accumulation, foam cell formation, and atherosclerosis. CONCLUSIONS: Our results suggest a potential role for CD36-mediated abnormal accumulations of oxidized lipids, such as 7-KCh, in air pollution-induced atherosclerosis progression.


Assuntos
Aorta/metabolismo , Doenças da Aorta/etiologia , Aterosclerose/etiologia , Antígenos CD36/sangue , Cetocolesteróis/sangue , Macrófagos/metabolismo , Material Particulado , Animais , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/patologia , Transporte Biológico , Antígenos CD36/deficiência , Antígenos CD36/genética , Modelos Animais de Doenças , Progressão da Doença , Células Espumosas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Exposição por Inalação , Masculino , Camundongos , Camundongos Knockout , Oxirredução , Tamanho da Partícula , Placa Aterosclerótica , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Tempo
6.
Magn Reson Med ; 71(1): 133-44, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23440705

RESUMO

PURPOSE: To describe and characterize a new approach to first-pass myocardial perfusion utilizing balanced steady-state free precession acquisition without the use of saturation recovery or other magnetization preparation. THEORY: The balanced steady-state free precession sequence is inherently sensitive to contrast agent enhancement of the myocardium. This sensitivity can be used to advantage in first-pass myocardial perfusion imaging by eliminating the need for magnetization preparation. METHODS: Bloch equation simulations, phantom experiments, and in vivo 2D imaging studies were run comparing the proposed technique with three other methods: saturation recovery spoiled gradient echo, saturation recovery steady-state free precession, and steady-state spoiled gradient echo without magnetization preparation. Additionally, an acquisition-reconstruction strategy for 3D perfusion imaging is proposed and initial experience with this approach is demonstrated in healthy subjects and one patient. RESULTS: Phantom experiments verified simulation results showing the sensitivity of the balanced steady-state free precession sequence to contrast agent enhancement in solid tissue is similar to that of magnetization-prepared acquisitions. Images acquired in normal volunteers showed the proposed technique provided superior signal and signal-to-noise ratio compared with all other sequences at baseline as well as postcontrast. CONCLUSIONS: A new approach to first-pass myocardial perfusion is presented that obviates the need for magnetization preparation and provides high signal-to-noise ratio.


Assuntos
Algoritmos , Vasos Coronários/anatomia & histologia , Interpretação de Imagem Assistida por Computador/métodos , Imageamento Tridimensional/métodos , Angiografia por Ressonância Magnética/métodos , Imagem de Perfusão do Miocárdio/métodos , Humanos , Aumento da Imagem/métodos , Valores de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
7.
Part Fibre Toxicol ; 11: 53, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25358444

RESUMO

BACKGROUND: Prior experimental and epidemiologic data support a link between exposure to fine ambient particulate matter (<2.5 µm in aerodynamic diameter, PM2.5) and development of insulin resistance/Type II diabetes mellitus (Type II DM). We investigated the role of hypothalamic inflammation in PM2.5-mediated diabetes development. METHODS: KKay mice, a genetically susceptible model of Type II DM, were assigned to either concentrated PM2.5 or filtered air (FA) for 4-8 weeks via a versatile aerosol concentrator and exposure system, or administered intra-cerebroventricular with either IKKß inhibitor (IMD-0354) or TNFα antibody (infliximab) for 4-5 weeks simultaneously with PM2.5 exposure. Glucose tolerance, insulin sensitivity, oxygen consumption and heat production were evaluated. At euthanasia, blood, spleen, visceral adipose tissue and hypothalamus were collected to measure inflammatory cells using flow cytometry. Standard immunohistochemical methods and quantitative PCR were used to assess targets of interest. RESULTS: PM2.5 exposure led to hyperglycemia and insulin resistance, which was accompanied by increased hypothalamic IL-6, TNFα, and IKKß mRNA expression and microglial/astrocyte reactivity. Targeting the NFκB pathway with intra-cerebroventricular administration of an IKKß inhibitor [IMD-0354, n = 8 for each group)], but not TNFα blockade with infliximab [(n = 6 for each group], improved glucose tolerance, insulin sensitivity, rectified energy homeostasis (O2 consumption, CO2 production, respiratory exchange ratio and heat generation) and reduced peripheral inflammation in response to PM2.5. CONCLUSIONS: Central inhibition of IKKß prevents PM2.5 mediated peripheral inflammation and exaggeration of type II diabetes. These results provide novel insights into how air pollution may mediate susceptibility to insulin resistance and Type II DM.


Assuntos
Benzamidas/farmacologia , Diabetes Mellitus Tipo 2/prevenção & controle , Hipotálamo/efeitos dos fármacos , Quinase I-kappa B/antagonistas & inibidores , Inflamação/prevenção & controle , Material Particulado/toxicidade , Inibidores de Proteínas Quinases/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Anticorpos Monoclonais/farmacologia , Benzamidas/administração & dosagem , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/imunologia , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Hipotálamo/enzimologia , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Inflamação/induzido quimicamente , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Infliximab , Exposição por Inalação/efeitos adversos , Injeções Intraventriculares , Insulina/sangue , Resistência à Insulina , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Consumo de Oxigênio/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , RNA Mensageiro/metabolismo , Medição de Risco , Termogênese/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
8.
Inhal Toxicol ; 26(1): 23-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24417404

RESUMO

CONTEXT: High-density lipoprotein (HDL) particles perform numerous vascular-protective functions. Animal studies demonstrate that exposure to fine or ultrafine particulate matter (PM) can promote HDL dysfunction. However, the impact of PM on humans remains unknown. OBJECTIVE: We aimed to determine the effect of exposure to coarse concentrated ambient particles (CAP) on several metrics of HDL function in healthy humans. METHODS: Thirty-two adults (25.9 ± 6.6 years) were exposed to coarse CAP [76.2 ± 51.5 µg·m(-3)] in a rural location and filtered air (FA) for 2 h in a randomized double-blind crossover study. Venous blood collected 2- and 20-h post-exposures was measured for HDL-mediated efflux of [(3)H]-cholesterol from cells and 20-h exposures for HDL anti-oxidant capacity by a fluorescent assay and paraoxonase activity. The changes [median (first, third quartiles)] between exposures among 29 subjects with available results were compared by matched Wilcoxon tests. RESULTS: HDL-mediated cholesterol efflux capacity did not differ between exposures at either time point [16.60% (15.17, 19.19) 2-h post-CAP versus 17.56% (13.43, 20.98) post-FA, p = 0.768 and 14.90% (12.47, 19.15) 20-h post-CAP versus 17.75% (13.22, 23.95) post-FA, p = 0.216]. HOI [0.26 (0.24, 0.35) versus 0.28 (0.25, 0.40), p = 0.198] and paraoxonase activity [0.54 (0.39, 0.82) versus 0.60 µmol·min(-1 )ml plasma(-1) (0.40, 0.85), p = 0.137] did not differ 20-h post-CAP versus FA, respectively. CONCLUSIONS: Brief inhalation of coarse PM from a rural location did not acutely impair several facets of HDL functionality. Whether coarse PM derived from urban sites, fine particles or longer term PM exposures can promote HDL dysfunction warrant future investigations.


Assuntos
Poluentes Atmosféricos/toxicidade , Lipoproteínas HDL/sangue , Material Particulado/toxicidade , Adolescente , Adulto , Poluição do Ar/efeitos adversos , Animais , Arildialquilfosfatase/sangue , Linhagem Celular Tumoral , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Lipoproteínas HDL/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Tamanho da Partícula , População Rural , Adulto Jovem
9.
Am Heart J ; 166(3): 392-400, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24016485

RESUMO

The goal of this systematic review was to assess the current understanding of the effects of exercise intervention on high-density lipoprotein (HDL) cholesterol (HDL-C) and changes in HDL function as well as modification of these effects by genomic factors. The reviewed studies demonstrate that exercise has modest effects on HDL-C with limited data suggesting an effect on HDL function. Genetic polymorphisms in proteins associated with HDL metabolism play a role in modifying the HDL-C response to exercise and possibly its function. Exercise as an intervention for patients at risk for cardiovascular events can lead to small improvements in HDL-C and potential changes in HDL function. There is an important modifier effect of genetics in determining these changes.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Exercício Físico/fisiologia , Lipoproteínas HDL/sangue , Lipoproteínas HDL/fisiologia , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/fisiopatologia , LDL-Colesterol/sangue , LDL-Colesterol/genética , LDL-Colesterol/fisiologia , Humanos , Lipoproteínas HDL/genética , Fatores de Risco
10.
Circ Res ; 108(6): 716-26, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21273555

RESUMO

RATIONALE: Chronic exposure to ambient air-borne particulate matter of < 2.5 µm (PM2.5) increases cardiovascular risk. The mechanisms by which inhaled ambient particles are sensed and how these effects are systemically transduced remain elusive. OBJECTIVE: To investigate the molecular mechanisms by which PM2.5 mediates inflammatory responses in a mouse model of chronic exposure. METHODS AND RESULTS: Here, we show that chronic exposure to ambient PM2.5 promotes Ly6C(high) inflammatory monocyte egress from bone-marrow and mediates their entry into tissue niches where they generate reactive oxygen species via NADPH oxidase. Toll-like receptor (TLR)4 and Nox2 (gp91(phox)) deficiency prevented monocyte NADPH oxidase activation in response to PM2.5 and was associated with restoration of systemic vascular dysfunction. TLR4 activation appeared to be a prerequisite for NAPDH oxidase activation as evidenced by reduced p47(phox) phosphorylation in TLR4 deficient animals. PM2.5 exposure markedly increased oxidized phospholipid derivatives of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine (oxPAPC) in bronchioalveolar lavage fluid. Correspondingly, exposure of bone marrow-derived macrophages to oxPAPC but not PAPC recapitulated effects of chronic PM2.5 exposure, whereas TLR4 deficiency attenuated this response. CONCLUSIONS: Taken together, our findings suggest that PM2.5 triggers an increase in oxidized phospholipids in lungs that then mediates a systemic cellular inflammatory response through TLR4/NADPH oxidase-dependent mechanisms.


Assuntos
NADPH Oxidases/metabolismo , Material Particulado/efeitos adversos , Receptor 4 Toll-Like/metabolismo , Doenças Vasculares/induzido quimicamente , Doenças Vasculares/etiologia , Administração por Inalação , Poluentes Atmosféricos/efeitos adversos , Animais , Exposição Ambiental , Ativação Enzimática , Inflamação/etiologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Oxirredução , Tamanho da Partícula , Material Particulado/administração & dosagem , Fosfolipídeos/metabolismo , Fatores de Tempo
11.
Arterioscler Thromb Vasc Biol ; 32(4): 962-70, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22308043

RESUMO

OBJECTIVE: Myeloid-related protein (Mrp) 8/14 complex (is a highly expressed extracellularly secreted protein, implicated in atherosclerosis. In this study, we evaluated the feasibility of targeting Mrp in vivo through synthetic immuno-nanoprobes. METHODS AND RESULTS: Anti-Mrp-14 and nonspecific IgG-conjugated gadolinium nanoprobes (aMrp-) were synthesized and characterized. Pharmacokinetics and vascular targeting via MRI of the formulations were assessed in vivo in high fat-fed apolipoprotein E deficient (ApoE(-/-)), ApoE(-/-)/Mrp14(-/-) (double knockout) and chow-fed wild-type (C57BL/6) mice. Bone marrow-derived myeloid progenitor cells were isolated from both ApoE(-/-) and double knockout mice, differentiated to macrophages, and were treated with LPS, with or without Mrp8, Mrp14, or Mrp8/14; conditioned media was used for in vitro studies. Mrp-activated cells secreted significant amounts of proinflammatory cytokines, which was abolished by pretreatment with aMrp-NP. We show in vitro that aMrp-NP binds endothelial cells previously treated with conditioned media containing Mrp8/14. MRI following intravenous delivery of aMrp-NP revealed prolonged and substantial delineation of plaque in ApoE(-/-) but not double knockout or wild-type animals. Nonspecific IgG-conjugated gadolinium nanoprobe-injected animals in all groups did not show vessel wall enhancement. Flow-cytometric analysis of aortic digesta revealed that aMrp-NP present in Ly-6G(+), CD11b(+), CD11c(+), and CD31(+) cells in ApoE(-/-) but not in double knockout animals. CONCLUSIONS: Targeted imaging with aMrp-NP demonstrates enhancement of plaque with binding to inflammatory cells and reduction in inflammation. This strategy has promise as a theranostic approach for atherosclerosis.


Assuntos
Albuminas/farmacocinética , Anti-Inflamatórios/farmacocinética , Anticorpos/metabolismo , Aterosclerose/metabolismo , Calgranulina A/imunologia , Calgranulina B/imunologia , Meios de Contraste/farmacocinética , Gadolínio DTPA/farmacocinética , Imunoconjugados/farmacocinética , Inflamação/metabolismo , Nanopartículas Metálicas , Albuminas/química , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Anticorpos/química , Anticorpos/farmacologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/diagnóstico , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/imunologia , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Células Cultivadas , Meios de Contraste/química , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Estudos de Viabilidade , Citometria de Fluxo , Gadolínio DTPA/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imunoconjugados/química , Inflamação/diagnóstico , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Progenitoras Mieloides/metabolismo , Distribuição Tecidual
12.
Part Fibre Toxicol ; 10: 43, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23968387

RESUMO

BACKGROUND: Inflammation and oxidative stress play critical roles in the pathogenesis of inhaled air pollutant-mediated metabolic disease. Inflammation in the adipose tissues niches are widely believed to exert important effects on organ dysfunction. Recent data from both human and animal models suggest a role for inflammation and oxidative stress in epicardial adipose tissue (EAT) as a risk factor for the development of cardiovascular disease. We hypothesized that inhalational exposure to concentrated ambient fine particulates (CAPs) and ozone (O3) exaggerates inflammation and oxidative stress in EAT and perirenal adipose tissue (PAT). METHODS: Eight- week-old Male Sprague-Dawley rats were fed a normal diet (ND) or high fructose diet (HFr) for 8 weeks, and then exposed to ambient AIR, CAPs at a mean of 356 µg/m3, O3 at 0.485 ppm, or CAPs (441 µg/m3) + O3 (0.497 ppm) in Dearborn, MI, 8 hours/day, 5 days/week, for 9 days over 2 weeks. RESULTS: EAT and PAT showed whitish color in gross, and less mitochondria, higher mRNA expression of white adipose specific and lower brown adipose specific genes than in brown adipose tissues. Exposure to CAPs and O3 resulted in the increase of macrophage infiltration in both EAT and PAT of HFr groups. Proinflammatory genes of Tnf-α, Mcp-1 and leptin were significantly upregulated while IL-10 and adiponectin, known as antiinflammatory genes, were reduced after the exposures. CAPs and O3 exposures also induced an increase in inducible nitric oxide synthase (iNOS) protein expression, and decrease in mitochondrial area in EAT and PAT. We also found significant increases in macrophages of HFr-O3 rats. The synergetic interaction of HFr and dirty air exposure on the inflammation was found in most of the experiments. Surprisingly, exposure to CAPs or O3 induced more significant inflammation and oxidative stress than co-exposure of CAPs and O3 in EAT and PAT. CONCLUSION: EAT and PAT are both white adipose tissues. Short-term exposure to CAPs and O3, especially with high fructose diet, induced inflammation and oxidative stress in EAT and PAT in rats. These findings may provide a link between air-pollution exposure and accelerated susceptibility to cardiovascular disease and metabolic complications.


Assuntos
Tecido Adiposo Branco/efeitos dos fármacos , Carboidratos da Dieta , Frutose , Exposição por Inalação/efeitos adversos , Ozônio/toxicidade , Paniculite/induzido quimicamente , Material Particulado/toxicidade , Adipocinas/genética , Adipocinas/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/ultraestrutura , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/ultraestrutura , Animais , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Rim , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Paniculite/genética , Paniculite/metabolismo , Paniculite/patologia , Pericárdio , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Medição de Risco , Fatores de Tempo
13.
Am J Hypertens ; 36(10): 542-550, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37439351

RESUMO

BACKGROUND: The resolution of inflammation is an active phenomenon important for switching off inflammatory processes once the harmful stimuli are removed and facilitate the return to homeostasis. Specialized pro-resolving mediators (SPMs), such as lipoxin A4, resolvin D1, and resolvin E1, derived from ω-3 or ω-6 polyunsaturated fatty acids, are crucial for the resolution of inflammation. We hypothesized that SPMs are decreased in hypertension which contributes to the acetylcholine-induced contraction in resistance arteries, which are well known to be mediated by leukotrienes and prostaglandins. Moreover, treatment with SPMs will decrease this contraction via formyl peptide receptor-2 (FPR-2) in resistance arteries from spontaneously hypertensive rats (SHR). METHODS AND RESULTS: We performed a comprehensive eicosanoid lipid panel analysis, and our data showed for the first time that precursors of SPMs are decreased in SHR, limiting the production of SPMs and resolution of inflammation in vivo. This phenomenon was associated with an increase in lipid peroxidation in resistance arteries. Although SPMs did not abolish acetylcholine-induced contraction, these lipid mediators improved endothelial function in arteries from SHR via FPR-2 activation at nanomolar concentrations. SPMs also buffered TNF-α-induced reactive oxygen species generation in endothelial cells from C57Bl/6 mice. CONCLUSIONS: We suggest that FPR-2 and SPMs could be revealed as a new target or therapeutic agent to improve vascular function in arteries from hypertensive rats.


Assuntos
Acetilcolina , Receptores de Formil Peptídeo , Animais , Camundongos , Ratos , Ácidos Docosa-Hexaenoicos/farmacologia , Células Endoteliais , Inflamação , Mediadores da Inflamação
14.
J Lipid Res ; 53(5): 879-887, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22355094

RESUMO

Although statin therapy is a cornerstone of current low density lipoprotein (LDL)-lowering strategies, there is a need for additional therapies to incrementally lower plasma LDL cholesterol. In this study, we investigated the effect of several methylenedioxyphenol derivatives in regulating LDL cholesterol through induction of LDL receptor (LDLR). INV-403, a modified methylenedioxyphenol derivative, increased LDLR mRNA and protein expression in HepG2 cells in a dose- and time-dependent fashion. These effects were apparent even under conditions of HMG-CoA reductase inhibition. Electrophoresis migration shift assays demonstrated that INV-403 activates SREBP2 but not SREBP1c, with immunoblot analysis showing an increased expression of the mature form of SREBP2. Knockdown of SREBP2 reduced the effect of INV-403 on LDLR expression. The activation of SREBP2 by INV-403 is partly mediated by Akt/GSK3ß pathways through inhibition of phosphorylation-dependent degradation by ubiquitin-proteosome pathway. Treatment of C57Bl/6j mice with INV-403 for two weeks increased hepatic SREBP2 levels (mature form) and upregulated LDLR with concomitant lowering of plasma LDL levels. Transient expression of a LDLR promoter-reporter construct, a SRE-mutant LDLR promoter construct, and a SRE-only construct in HepG2 cells revealed an effect predominantly through a SRE-dependent mechanism. INV-403 lowered plasma LDL cholesterol levels through LDLR upregulation. These results indicate a role for small molecule approaches other than statins for lowering LDL cholesterol.


Assuntos
Anticolesterolemiantes/química , Anticolesterolemiantes/farmacologia , LDL-Colesterol/sangue , Fenóis/química , Fenóis/farmacologia , Receptores de LDL/genética , Ativação Transcricional/efeitos dos fármacos , Animais , Células Hep G2 , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética
15.
Circulation ; 124(21): 2338-49, 2011 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22007077

RESUMO

BACKGROUND: Dipeptidyl-peptidase 4 (DPP-4) inhibitors are increasingly used to accomplish glycemic targets in patients with type II diabetes mellitus. Because DPP-4 is expressed in inflammatory cells, we hypothesized that its inhibition will exert favorable effects in atherosclerosis. METHODS AND RESULTS: Male LDLR(-/-) mice (6 weeks) were fed a high-fat diet or normal chow diet for 4 weeks and then randomized to vehicle or alogliptin, a high-affinity DPP-4 inhibitor (40 mg · kg(-1) · d(-1)), for 12 weeks. Metabolic parameters, blood pressure, vascular function, atherosclerosis burden, and indexes of inflammation were obtained in target tissues, including the vasculature, adipose, and bone marrow, with assessment of global and cell-specific inflammatory pathways. In vitro and in vivo assays of DPP-4 inhibition (DPP-4i) on monocyte activation/migration were conducted in both human and murine cells and in a short-term ApoE(-/-) mouse model. DPP-4i improved markers of insulin resistance and reduced blood pressure. DPP-4i reduced visceral adipose tissue macrophage content (adipose tissue macrophages; CD11b(+), CD11c(+), Ly6C(hi)) concomitant with upregulation of CD163. DPP-4 was highly expressed in bone marrow-derived CD11b(+) cells, with DPP-4i downregulating proinflammatory genes in these cells. DPP-4i decreased aortic plaque with a striking reduction in plaque macrophages. DPP-4i prevented monocyte migration and actin polymerization in in vitro assays via Rac-dependent mechanisms and prevented in vivo migration of labeled monocytes to the aorta in response to exogenous tumor necrosis factor-α and DPP-4. CONCLUSION: DPP-4i exerts antiatherosclerotic effects and reduces inflammation via inhibition of monocyte activation/chemotaxis. These findings have important implications for the use of this class of drugs in atherosclerosis.


Assuntos
Aterosclerose/patologia , Aterosclerose/prevenção & controle , Quimiotaxia/fisiologia , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Inflamação/patologia , Inflamação/prevenção & controle , Monócitos/patologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Movimento Celular/fisiologia , Quimiotaxia/efeitos dos fármacos , Inibidores da Dipeptidil Peptidase IV/farmacologia , Modelos Animais de Doenças , Glucose/metabolismo , Inflamação/fisiopatologia , Resistência à Insulina/fisiologia , Masculino , Metabolismo/efeitos dos fármacos , Camundongos , Camundongos Knockout , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Tempo , Uracila/análogos & derivados , Uracila/farmacologia , Uracila/uso terapêutico
16.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L399-409, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22160305

RESUMO

The purpose of this study was to investigate the effects of chronically inhaled particulate matter <2.5 µm (PM(2.5)) on inflammatory cell populations in the lung and systemic circulation. A prominent component of air pollution exposure is a systemic inflammatory response that may exaggerate chronic diseases such as atherosclerosis and insulin resistance. T cell response was measured in wild-type C57B/L6, Foxp3-green fluorescent protein (GFP) "knockin," and chemokine receptor 3 knockout (CXCR3(-/-)) mice following 24-28 wk of PM(2.5) or filtered air. Chronic PM(2.5) exposure resulted in increased CXCR3-expressing CD4(+) and CD8(+) T cells in the lungs, spleen, and blood with elevation in CD11c(+) macrophages in the lung and oxidized derivatives of 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine in wild-type mice. CXCR3 deficiency decreased T cells in the lung. GFP(+) regulatory T cells increased with PM(2.5) exposure in the spleen and blood of Foxp3-GFP mice but were present at very low levels in the lung irrespective of PM(2.5) exposure. Mixed lymphocyte cultures using primary, PM(2.5)-treated macrophages demonstrated enhanced T cell proliferation. Our experiments indicate that PM(2.5) potentiates a proinflammatory Th1 response involving increased homing of CXCR3(+) T effector cells to the lung and modulation of systemic T cell populations.


Assuntos
Poluição do Ar , Ativação Linfocitária , Material Particulado/toxicidade , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Contagem de Células , Proliferação de Células , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Expressão Gênica , Imunidade Inata , Interleucina-17/genética , Interleucina-17/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Linfonodos/patologia , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Material Particulado/imunologia , Fosfolipídeos/metabolismo , Regiões Promotoras Genéticas , Receptores CXCR3/deficiência , Receptores CXCR3/genética , Linfócitos T Reguladores
17.
Part Fibre Toxicol ; 9: 40, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23126276

RESUMO

BACKGROUND: It has been well recognized that toxicity of fine ambient air particulate matter (PM(2.5)) may depend on its chemical constituents, including components such as soluble metals that may theoretically exert distinctive effects. We have recently demonstrated an important effect of PM(2.5) on metabolic function. Since transition metals, such as nickel (Ni), represent an important component of exposure in certain environments, and may significantly influence the toxicity of inhalational exposure, we investigated the effects of Ni as a variable component of ambient PM(2.5) exposure. METHODS: Male ApoE knockout mice were exposed to filtered air (FA), fine-sized nickel sulfate particles alone (Ni) at 0.44 µg/m(3), concentrated ambient air PM(2.5) (CAPs) at a mean of 70 µg/m(3), or CAPs+Ni in Tuxedo, NY, 6 hours/day, 5 days/week, for 3 months. RESULTS: Exposure to Ni, irrespective of co-exposure to CAPs, resulted in body weight gain, while exposure to CAPs+Ni significantly enhanced fasting glucose and worsened insulin resistance measures (HOMA-IR), when compared with exposure to CAPs alone. CAPs+Ni exposure induced a significant decrease in phosphorylation of AMP-activated protein kinase (AMPK) α. Exposure to Ni or CAPs+Ni significantly induced microcirculatory dysfunction and increased monocytic cell infiltration into lung and adipose, and decreased uncoupling protein 1 expression at gene and protein levels and several brown adipocyte-specific genes in adipose tissue. CONCLUSIONS: Ni exposure has effects on metabolic and inflammatory parameters that are comparable to that of CAPs. Additionally, Ni synergistically exacerbates CAPs-induced adverse effects on some of, but not all of, these parameters, that may be mediated via the AMPK signaling pathway. These findings have important implications for inhaled transition metal toxicity that may exert synergistic effects with other PM(2.5) components.


Assuntos
Poluentes Atmosféricos/toxicidade , Exposição por Inalação/efeitos adversos , Resistência à Insulina , Mitocôndrias/efeitos dos fármacos , Níquel/toxicidade , Material Particulado/toxicidade , Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/efeitos dos fármacos , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Apolipoproteínas E/genética , Glicemia/análise , Citocinas/sangue , Sinergismo Farmacológico , Teste de Tolerância a Glucose , Resistência à Insulina/imunologia , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/genética , Tamanho Mitocondrial/efeitos dos fármacos , Tamanho da Partícula , Fosforilação , Reação em Cadeia da Polimerase em Tempo Real , Proteína Desacopladora 1
18.
Theranostics ; 12(6): 2741-2757, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35401813

RESUMO

Background: Glucagon-like peptide-1 receptor (GLP-1R) agonists are powerful glycemia-lowering agents, which have systematically been shown to lower cardiovascular events and mortality. These beneficial effects were difficult to pinpoint within atherosclerotic plaque due to lack of particular specificity of such agonists to the vascular cells and an inadequate understanding of the GLP-1R expression in atherosclerosis. Here, we hypothesized that the direct engagement of the GLP-1R in atherosclerosis by targeted agonists will alleviate vascular inflammation and plaque burden, even at a very low dose. Methods: The expression of GLP-1 receptor (GLP-1R, Glp1r mRNA) in human lesions with pathologic intimal thickening, Apoe-/- mouse atheroma and cultured immune/non-immune cells was investigated using genetic lineage tracing, Southern blotting and validated antisera against human GLP-1R. Protease-resistant and "activatable" nanoparticles (NPs) carrying GLP-1R agonist liraglutide (GlpNP) were engineered and synthesized. Inclusion of gadolinium chelates into GlpNP allowed for imaging by MRI. Atherosclerotic Apoe-/- mice were treated intravenously with a single dose (30 µg/kg of liraglutide) or chronically (1 µg/kg, 6 weeks, 2x/week) with GlpNP, liraglutide or control NPs, followed by assessment of metabolic parameters, atheroma burden, inflammation and vascular function. Results: Humal plaque specimens expressed high levels of GLP-1R within the locus of de-differentiated smooth muscle cells that also expressed myeloid marker CD68. However, innate immune cells under a variety of conditions expressed very low levels of Glp1r, as seen in lineage tracing and Southern blotting experiments examining full-length open reading frame mRNA transcripts. Importantly, de-differentiated vascular smooth muscle cells demonstrated significant Glp1r expression levels, suggesting that these could represent the cells with predominant Glp1r-positivity in atherosclerosis. GlpNP resisted proteolysis and demonstrated biological activity including in vivo glycemia lowering at 30 µg/kg and in vitro cholesterol efflux. Activatable properties of GlpNP were confirmed in vitro by imaging cytometry and in vivo using whole organ imaging. GlpNP targeted CD11b+/CD11c+ cells in circulation and smooth muscle cells in aortic plaque in Apoe-/- mice when assessed by MRI and fluorescence imaging. At a very low dose of 1 µg/kg, previously known to have little effect on glycemia and weight loss, GlpNP delivered i.v. for six weeks reduced triglyceride-rich lipoproteins in plasma, plaque burden and plaque cholesterol without significant effects on weight, glycemia and plasma cholesterol levels. Conclusions: GlpNP improves atherosclerosis at weight-neutral doses as low as 1 µg/kg with the effects independent from the pancreas or the central nervous system. Our study underlines the importance of direct actions of GLP-1 analogs on atherosclerosis, involving cholesterol efflux and inflammation. Our findings are the first to suggest the therapeutic modulation of vascular targets by GlpNP, especially in the context of smooth muscle cell inflammation.


Assuntos
Aterosclerose , Receptor do Peptídeo Semelhante ao Glucagon 1 , Placa Aterosclerótica , Animais , Aterosclerose/diagnóstico por imagem , Aterosclerose/tratamento farmacológico , Diferenciação Celular , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Inflamação/metabolismo , Liraglutida/farmacologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Músculo Liso/metabolismo , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/tratamento farmacológico , Proteólise , RNA Mensageiro/metabolismo
19.
Sci Transl Med ; 14(660): eabj7465, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-36044595

RESUMO

Arterial and venous thrombosis constitutes a major source of morbidity and mortality worldwide. Long considered as distinct entities, accumulating evidence indicates that arterial and venous thrombosis can occur in the same populations, suggesting that common mechanisms are likely operative. Although hyperactivation of the immune system is a common forerunner to the genesis of thrombotic events in both vascular systems, the key molecular control points remain poorly understood. Consequently, antithrombotic therapies targeting the immune system for therapeutics gain are lacking. Here, we show that neutrophils are key effectors of both arterial and venous thrombosis and can be targeted through immunoregulatory nanoparticles. Using antiphospholipid antibody syndrome (APS) as a model for arterial and venous thrombosis, we identified the transcription factor Krüppel-like factor 2 (KLF2) as a key regulator of neutrophil activation. Upon activation through genetic loss of KLF2 or administration of antiphospholipid antibodies, neutrophils clustered P-selectin glycoprotein ligand 1 (PSGL-1) by cortical actin remodeling, thereby increasing adhesion potential at sites of thrombosis. Targeting clustered PSGL-1 using nanoparticles attenuated neutrophil-mediated thrombosis in APS and KLF2 knockout models, illustrating the importance and feasibility of targeting activated neutrophils to prevent pathological thrombosis. Together, our results demonstrate a role for activated neutrophils in both arterial and venous thrombosis and identify key molecular events that serve as potential targets for therapeutics against diverse causes of immunothrombosis.


Assuntos
Síndrome Antifosfolipídica , Trombose , Trombose Venosa , Anticorpos Antifosfolipídeos , Humanos , Neutrófilos/metabolismo , Trombose/etiologia
20.
Drug Deliv ; 28(1): 408-421, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33594923

RESUMO

Native nanostructured lipoproteins such as low- and high-density lipoproteins (LDL and HDL) are powerful tools for the targeted delivery of drugs and imaging agents. While the cellular recognition of well-known HDL-based carriers occurs via interactions with an HDL receptor, the selective delivery and uptake of LDL particles by target cells are more complex. The most well-known mode of LDL-based delivery is via the interaction between apolipoprotein B (Apo-B) - the main protein of LDL - and the low-density lipoprotein receptor (LDLR). LDLR is expressed in the liver, adipocytes, and macrophages, and thus selectively delivers LDL carriers to these cells and tissues. Moreover, the elevated expression of LDLR in tumor cells indicates a role for LDL in the targeted delivery of chemotherapy drugs. In addition, chronic inflammation associated with hypercholesterolemia (i.e., high levels of endogenous LDL) can be abated by LDL carriers, which outcompete the deleterious oxidized LDL for uptake by macrophages. In this case, synthetic LDL nanocarriers act as 'eat-me' signals and exploit mechanisms of native LDL uptake for targeted drug delivery and imaging. Lastly, recent studies have shown that the delivery of LDL-based nanocarriers to macrophages via fluid-phase pinocytosis is a promising tool for atherosclerosis imaging. Hence, the present review summarizes the use of natural and synthetic LDL-based carriers for drug delivery and imaging and discusses various mechanisms of targeting.


Assuntos
Lipoproteínas LDL/química , Lipoproteínas LDL/farmacologia , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Portadores de Fármacos/química , Humanos , Hipercolesterolemia/tratamento farmacológico , Hipercolesterolemia/metabolismo , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanomedicina/métodos , Receptores de Lipoproteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA