Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Circ Res ; 101(11): 1130-8, 2007 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-17901362

RESUMO

Adenosine is a proangiogenic purine nucleoside released from ischemic and hypoxic tissues. Of the 4 adenosine receptor (AR) subtypes (A1, A2A, A2B, and A3), the A2 and A3 have been previously linked to the modulation of angiogenesis. We used the chicken chorioallantoic membrane (CAM) model to determine whether A1 AR activation affects angiogenesis. We cloned and pharmacologically characterized chicken AR subtypes to evaluate the selectivity of various agonists and antagonists. Application of the A1 AR-selective agonist N6-cyclopentyladenosine (CPA; 100 nmol/L) to the CAM resulted in a 40% increase in blood vessel number (P<0.01), which was blocked by the A1 AR-selective antagonist C8-(N-methylisopropyl)-amino-N6-(5'-endohydroxy)-endonorbornan-2-yl-9-methyladenine (WRC-0571; 1 micromol/L). Selective A2A AR agonists did not stimulate angiogenesis in the CAM. In an ex vivo rat aortic ring model of angiogenesis that includes cocultured endothelial cells, fibroblasts, and smooth muscle cells, 50 nmol/L CPA did not directly stimulate capillary formation; however, medium from human mononuclear cells pretreated with CPA, but not vehicle, increased capillary formation by 48% (P<0.05). This effect was blocked by WRC-0571 (1.5 micromol/L) or anti-VEGF antibody (1 microg/mL). CPA (5 nmol/L) stimulated a 1.7-fold increase in VEGF release from the mononuclear cells. This is the first study to show that A1 AR activation induces angiogenesis. Stimulation of A2 ARs on endothelial cells results in proliferation and tube formation, and A2 and A3 ARs on inflammatory cells modulate release of angiogenic factors. We conclude that adenosine promotes a coordinated angiogenic response through its interactions with multiple receptors on multiple cell types.


Assuntos
Monócitos/metabolismo , Neovascularização Fisiológica , Receptor A1 de Adenosina/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina , Animais , Aorta , Embrião de Galinha , Humanos , Ratos , Receptor A1 de Adenosina/metabolismo , Receptor A3 de Adenosina/metabolismo , Receptor A3 de Adenosina/fisiologia , Receptores A2 de Adenosina/metabolismo , Receptores A2 de Adenosina/fisiologia
2.
Biochem J ; 391(Pt 3): 575-80, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16022683

RESUMO

The A(2A) adenosine receptor (A(2A)AR) mediates anti-inflammatory actions of adenosine in a variety of cell types. LPS (lipopolysaccharide) was reported to induce a small (<2-fold) increase in the expression of A(2A)AR mRNA in human monocytes and monocytic cell lines. We investigated the effects of LPS on the expression of adenosine receptor mRNAs in primary mouse IPMPhi (intraperitoneal macrophages), human macrophages and Wehi-3 cells. Treatment with 10 ng/ml LPS for 4 h produced a >100-fold increase in A(2A)AR mRNA. LPS-induced increases in mRNA for A(2A)AR and TNFalpha (tumour necrosis factor alpha) are reduced by 90% in IPMPhi pretreated with the NF-kappaB (nuclear factor kappaB) inhibitor, BAY 11-7082 {(E)3-[(4-methylphenyl)sulphonyl]-2-propenenitrile; 10 microM}. In Wehi-3 cells exposed to LPS, A(2A)AR and A(2B)AR transcripts are elevated by 290- and 10-fold respectively, the A(1)AR transcript is unchanged and the A(3)AR transcript is decreased by 67%. The induction of A(2A)AR mRNA by LPS is detectable after 1 h, reaches a peak at 6 h at 600 times control and remains elevated beyond 24 h. The ED50 (effective dose) of LPS is 2.3 ng/ml. A(2A)AR receptor number, measured by 125I-ZM241385 binding to whole cells, is undetectable in naïve cells and increases linearly at a rate of 23 receptors x cell(-1) x min(-1) to a B(max) of 348 fmol/mg (28000 receptors/cell) in 20 h. The increase in receptor number is correlated with an increase in the potency of an A(2A) agonist (4-{3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl}-cyclohexanecarboxylic acid methyl ester; referred to as ATL146e) to stimulate cAMP in these cells. After LPS pretreatment, the potency of the A(2A) agonist, ATL146e, to reduce TNFalpha release from IPMPhi was increased by 200-fold. The results support the hypothesis that regulation of adenosine receptor expression, especially up-regulation of the A(2A)AR, is part of a delayed feedback mechanism initiated through NF-kappaB to terminate the activation of human and mouse macrophages.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , NF-kappa B/metabolismo , Receptor A2A de Adenosina/genética , Transcrição Gênica/efeitos dos fármacos , Animais , Células Cultivadas , Ácidos Cicloexanocarboxílicos/farmacologia , Humanos , Macrófagos/metabolismo , Camundongos , Nitrilas/farmacologia , Ligação Proteica , Purinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sulfonas/farmacologia , Triazinas/farmacologia , Triazóis/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
3.
Circulation ; 106(5): 592-8, 2002 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-12147542

RESUMO

BACKGROUND: 99mTc-RP517 is a new leukotriene B4 (LTB4) receptor antagonist developed for imaging acute inflammation or infection. A unique property of 99mTc-RP517 is its ability to label white blood cells in vivo after intravenous injection. The goals of this study were to determine relative 99mTc-RP517 binding to human leukocyte subtypes and the 99mTc-RP517 uptake pattern in canine myocardium where inflammation was induced by either coronary occlusion and reperfusion or tumor necrosis factor alpha (TNFalpha) injection. METHODS AND RESULTS: Fluorescence-activated cell sorter analysis was performed on whole human blood (n=2) and isolated neutrophils (n= 4) with a fluorescent analog of 99mTc-RP517, [F]-RP517. In whole blood, [F]-RP517 (500 nmol/L) preferentially labeled neutrophils. On isolated neutrophils, [F]-RP517 (10 nmol/L) binding was inhibited by 44% when LTB4 (400 nmol/L) was added. 99mTc-RP517 was injected intravenously in anesthetized, open-chest dogs before coronary occlusion (90 minutes) and reperfusion (120 minutes) (n=9) or before intramyocardial TNFalpha injection (n=3). Ex vivo images of heart slices were acquired. The left ventricle was divided into 72 segments for flow and 99mTc-RP517 uptake analysis. There was an inverse exponential relationship between 99mTc-RP517 uptake and occlusion flow (r=0.73). In the same 15 segments, 99mTc-RP517 uptake was highly correlated with the neutrophil enzyme myeloperoxidase (r=0.91). Ex vivo images revealed tracer uptake in the reperfused area (ischemic to normal count ratio=2.7+/-0.2). CONCLUSIONS: RP517 binds to the neutrophil LTB4 receptor after intravenous injection. After reperfusion, 99mTc-RP517 uptake correlated with myeloperoxidase and was observed on ex vivo images, indicating that this tracer may have potential as an inflammation-imaging agent.


Assuntos
Doença das Coronárias/fisiopatologia , Inflamação/diagnóstico , Miocardite/diagnóstico , Neutrófilos/metabolismo , Compostos de Organotecnécio , Receptores do Leucotrieno B4/antagonistas & inibidores , Animais , Autorradiografia , Ligação Competitiva , Circulação Coronária , Cães , Vias de Administração de Medicamentos , Citometria de Fluxo , Hemodinâmica , Humanos , Inflamação/induzido quimicamente , Injeções , Injeções Intravenosas , Reperfusão Miocárdica , Miocardite/induzido quimicamente , Miocárdio/imunologia , Miocárdio/metabolismo , Neutrófilos/citologia , Neutrófilos/imunologia , Compostos de Organotecnécio/administração & dosagem , Compostos de Organotecnécio/farmacocinética , Peroxidase/metabolismo , Valor Preditivo dos Testes , Receptores do Leucotrieno B4/metabolismo , Traumatismo por Reperfusão/fisiopatologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/farmacologia
4.
J Leukoc Biol ; 75(1): 127-34, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14525968

RESUMO

The alpha 4/beta 1 integrin very late antigen-4 (CD49d/CD29) is up-regulated on circulating neutrophils of septic patients. Although no individual agent mimics this effect of sepsis, we now report that following priming of human neutrophils with lipopolysaccharide or tumor necrosis factor alpha (TNF-alpha), addition of formyl-Met-Leu-Phe (fMLP) results in a "stimulated", sepsis-like, four- to fivefold rise in CD49d expression. TNF/fMLP stimulation also produced a similar increase in CD49d-mediated adhesion of neutrophils to a vascular cell adhesion molecule-1 (VCAM-1)-coated surface. Adenosine is a naturally occurring, anti-inflammatory mediator released from injured or inflamed tissues. We observed that stimulated neutrophil CD49d expression was decreased by activation of A(2A) adenosine receptors (A(2A)AR) with the selective agonist 4-[3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylicacid methyl ester (ATL146e; EC(50)=6.4 nM). ATL146e (100 nM) also reduced the fraction of stimulated neutrophils that adhered to VCAM-1 from 38 +/- 6% to 27 +/- 5%. Inhibition of CD49d expression was equally inhibited by ATL146e, added before or after TNF priming, and was reversed by incubation with the A(2A)AR-selective antagonist 4-[2-[7-amino-2-(2-furyl) (1, 2, 4)triazolo(2,3-a) (1, 3, 5)triazin-5-yl-amino]ethyl]-phenol (ZM241385; 100 nM). A suboptimal ATL146e concentration (1 nM) combined with the type IV phosphodiesterase inhibitor rolipram (100 nM) synergistically decreased stimulated CD49d expression by >50%. The cyclic adenosine monophosphate (cAMP)-dependent kinase [protein kinase A (PKA)] inhibitor H-89 (10 microM) reversed the effect of ATL146e on stimulated CD49d expression. Other means of increasing cAMP in neutrophils also decreased stimulated CD49d expression. We conclude that adenosine binding to A(2A)AR counteracts stimulation of neutrophil CD49d integrin expression and neutrophil binding to VCAM-1 via a cAMP/PKA-mediated pathway.


Assuntos
Integrina alfa4beta1/genética , Ativação de Neutrófilo/imunologia , Neutrófilos/imunologia , Receptor A2A de Adenosina/fisiologia , Adenosina/fisiologia , Antígenos CD/sangue , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Humanos , Técnicas In Vitro , Integrina alfa4/sangue , Integrina alfa4beta1/efeitos dos fármacos , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Receptor A2A de Adenosina/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Fator de Necrose Tumoral alfa/farmacologia
5.
Curr Opin Investig Drugs ; 4(11): 1313-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14758770

RESUMO

Stressed or injured tissues release endogenous adenosine, which blocks potentially destructive inflammatory cascades by binding to A2A adenosine receptors (ARs) and decreasing activation of platelets, leukocytes and endothelial cells. In these tissues, adenosine acts by reducing expression of adhesion molecules and release of pro-inflammatory mediators (e.g., reactive oxygen species, elastase and tumor necrosis factor-alpha). Synthetic A2A selective AR agonists are currently undergoing preclinical testing for the treatment of allergen-induced inflammation, ischemia-reperfusion injury, sepsis and autoimmune diseases. This review discusses potential disease targets for A2A AR agonist treatment, mechanisms of A2A AR agonist action and considerations for synthetic A2A AR agonist design.


Assuntos
Agonistas do Receptor A2 de Adenosina , Adenosina , Anti-Inflamatórios não Esteroides/farmacologia , Inflamação/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacologia , Animais , Anti-Inflamatórios não Esteroides/química , Humanos , Inflamação/tratamento farmacológico , Inflamação/etiologia , Estrutura Molecular
6.
Am J Respir Crit Care Med ; 175(10): 1027-35, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17322107

RESUMO

RATIONALE: Excessive recruitment of polymorphonuclear leukocytes (PMNs) to the lung promotes acute lung injury (ALI). Chemokine receptors and adhesion molecules initiate leukocyte-endothelial interactions, but mediators of PMN migration through the alveolo-capillary membrane remain to be identified. p21-Activated kinase (PAK) is an effector of small GTPases and has been implicated in cell migration. OBJECTIVES: To test the role of PAK in ALI. METHODS: An inhibitory PAK peptide was used to determine the role of PAK in cytoskeletal actin polymerization, cell adhesion, and oxidative burst. PMN migration was investigated in vitro and in a murine model of lipopolysaccharide-induced lung injury. MEASUREMENTS AND MAIN RESULTS: PMN migration into lung interstitium and alveolar space was suppressed by an inhibitory PAK peptide. Neutrophils that had taken up the inhibitory PAK peptide were unable to enter the alveolar space. CXCL2/3, an important PMN chemoattractant in murine lung injury, induced PAK phosphorylation in PMNs. Blocking PAK function inhibited chemotaxis, chemokine-induced cytoskeletal actin polymerization, and adhesion-induced oxidative burst. CONCLUSIONS: We conclude that neutrophil PAK is a critical mediator of PMN migration and may be an attractive target in ALI.


Assuntos
Quimiotaxia de Leucócito , Neutrófilos/imunologia , Proteínas Serina-Treonina Quinases/fisiologia , Síndrome do Desconforto Respiratório/imunologia , Actinas/química , Animais , Adesão Celular/efeitos dos fármacos , Quimiocinas CXC/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Camundongos , Neutrófilos/enzimologia , Neutrófilos/ultraestrutura , Peptídeos/farmacologia , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Interleucina-8B/metabolismo , Explosão Respiratória , Quinases Ativadas por p21
7.
Bioorg Med Chem ; 14(11): 3654-61, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16458010

RESUMO

During the search for second-generation adenosine A(1) receptor antagonist alternatives to the clinical candidate 8-(3-oxa-tricyclo[3.2.1.0(2,4)]oct-6-yl)-1,3-dipropyl-3,7-dihydro-purine-2,6-dione (BG9719), we developed a series of novel xanthines substituted with norbornyl-lactones that possessed high binding affinities for adenosine A(1) receptors and in vivo activity.


Assuntos
Antagonistas do Receptor A1 de Adenosina , Compostos Bicíclicos com Pontes/química , Lactonas/química , Norbornanos/farmacologia , Xantinas/farmacologia , Antagonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A3 de Adenosina , Animais , Ligação Competitiva/efeitos dos fármacos , Linhagem Celular , Humanos , Ligantes , Estrutura Molecular , Norbornanos/síntese química , Norbornanos/química , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Xantinas/síntese química , Xantinas/química
8.
Expert Opin Investig Drugs ; 14(7): 797-806, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16022569

RESUMO

Extracellular adenosine binds specifically to a family of four G protein-coupled cell-surface adenosine receptors (ARs). As the activation of the A2AAR modulates the activity of multiple inflammatory cells including neutrophils, macrophages and T lymphocytes, the receptor is considered to be a promising pharmacological target for the treatment of inflammatory disorders. Although adenosine binds nonselectively to all four AR subtypes, A2AAR selective agonists have been developed and shown to inhibit multiple manifestations of inflammatory cell activation including superoxide anion generation, cytokine production and adhesion molecule expression. A2AAR agonists are also vasodilators, but the inhibition of inflammation occurs at low doses that produce few or no cardiovascular side effects. Therefore, the selective activation of the A2AAR by these compounds holds significant potential in the treatment of inflammation.


Assuntos
Agonistas do Receptor A2 de Adenosina , Anti-Inflamatórios não Esteroides/uso terapêutico , Drogas em Investigação/uso terapêutico , Tecnologia Farmacêutica/tendências , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Drogas em Investigação/química , Drogas em Investigação/farmacologia , Humanos , Receptor A2A de Adenosina/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 288(4): H1851-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15591104

RESUMO

Adenosine and adenosine A(2A) receptor agonists have been shown to limit myocardial infarct size when given at vasodilatory doses during reperfusion. This beneficial effect is thought to be due, in part, to stimulation of adenosine A(2A) receptors on inflammatory cells. The specific aims of this study were to determine whether the anti-inflammatory and cardioprotective properties of a novel adenosine A(2A) receptor agonist, ATL-146e (ATL), alone or in combination with the phosphodiesterase IV inhibitor rolipram would occur using very low, nonvasodilating doses. In a canine model of reperfused myocardial infarction, low-dose ATL given alone reduced infarct size by 45% (P < 0.05 vs. control). When ATL was combined with a very low dose of rolipram (0.001 microg.kg(-1).min(-1)), a marked reduction in P-selectin expression and neutrophil infiltration (51% lower; P < 0.001 vs. control) was seen and the infarct size reduction (58% lower; P < 0.01 vs. control) was greater than observed with ATL (45% lower; P < 0.05) or rolipram (33% lower; P < 0.05) alone. In conclusion, a low, nonvasodilating dose of ATL, a highly selective adenosine A(2A) receptor agonist, reduced infarct size after reperfusion. Furthermore, combining ATL and the phosphodiesterase IV inhibitor rolipram reduced infarct size even more than either agent alone. Such combination therapy may be beneficial clinically by potentiating cardioprotection after coronary reperfusion at doses far below those producing vasodilatation or side effects.


Assuntos
Agonistas do Receptor A2 de Adenosina , Ácidos Cicloexanocarboxílicos/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Purinas/farmacologia , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Animais , Cardiotônicos/farmacologia , Circulação Coronária/efeitos dos fármacos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Cães , Quimioterapia Combinada , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Miocardite/tratamento farmacológico , Miocardite/imunologia , Miocardite/patologia , Inibidores de Fosfodiesterase/farmacologia , Explosão Respiratória/efeitos dos fármacos , Rolipram/farmacologia
10.
J Infect Dis ; 189(10): 1897-904, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15122527

RESUMO

BACKGROUND: Sepsis is currently treated with antibiotics and various adjunctive therapies that are not very effective. METHODS: Mouse survival (4-5 days) and peritoneal and blood bacteria counts were determined after challenge with intraperitoneal lipopolysaccharide (LPS) or live Escherichia coli. RESULTS: The A(2A) adenosine receptor (AR) agonist 4-[3-[6-amino-9-(5-ethylcarbamoyl-3, 4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylic acid methyl ester (ATL146e; 0.05-50 mu g/kg) protected mice from challenge with LPS, and protection occurred when treatment was delayed up to 24 h after challenge. Deletion of the A (2A) AR gene, Adora2a, inhibited protection by ATL146e. A putative A (3)AR agonist, N(6)-3-iodobenzyladenosine-5'-N-methyluronamide (IB-MECA; 500 mu g/kg but not 5 or 50 mu g/kg) protected mice from challenge with LPS. The protective effects of both ATL146e and IB-MECA were counteracted by the A(2A) AR selective antagonist 4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo[2,3-a][1,3,5]triazin-5-yl-amino]ethyl)-phenol. In the live E. coli model, treatment with ATL146e (50 mu g/kg initiated 8 h after infection) increased survival in mice treated with ceftriaxone (5 days) from 40% to 100%. Treatment with ATL146e did not affect peritoneal numbers of live E. coli at the time of death or 120 h after infection but did increase numbers of peritoneal neutrophils and decreased the number of live E. coli in blood. CONCLUSIONS: AR agonists increase mouse survival in endotoxemia and sepsis via A(2A) AR-mediated mechanisms and reduce the number of live bacteria in blood.


Assuntos
Adenosina/análogos & derivados , Ácidos Cicloexanocarboxílicos/farmacologia , Endotoxemia/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Purinas/farmacologia , Receptores A2 de Adenosina/imunologia , Sepse/imunologia , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina , Animais , Modelos Animais de Doenças , Feminino , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sobrevida
11.
J Infect Dis ; 185(9): 1297-306, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12001047

RESUMO

The effects of purified toxin A in vitro on the shape and function of polymorphonuclear leukocytes (PMNL) were examined. Toxin A induced changes in adherent PMNL shape from a compact spherical or pyramidal shape to a thin and rope-like shape. This change in shape was accompanied by rearrangement of the F-actin cytoskeleton into aggregates. Toxin A-treated PMNL exhibited increased adherence and expressed less L-selectin and more Mac-1, compared with untreated PMNL. In contrast to these proinflammatory actions, toxin A impaired both directed and non-directed PMNL migration in response to N-formylmethionylleucylphenylalanine. In addition, toxin A decreased the oxidative activity of adherent PMNL stimulated by recombinant human tumor necrosis factor-alpha. These effects could be explained by toxin A-induced glucosylation of the signaling small-size guanine 5'-triphosphate-binding proteins of the Rho family in human PMNL.


Assuntos
Toxinas Bacterianas/toxicidade , Enterotoxinas/toxicidade , Neutrófilos/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Citocalasina B/farmacologia , Citoesqueleto/efeitos dos fármacos , Relação Dose-Resposta a Droga , Glicosilação , Humanos , Antígeno de Macrófago 1/biossíntese , Microscopia Eletrônica de Varredura , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Neutrófilos/fisiologia , Neutrófilos/ultraestrutura , Fator de Necrose Tumoral alfa/farmacologia , Proteínas rho de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa