Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Eur Heart J ; 36(17): 1049-58, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24553721

RESUMO

AIMS: There is a need for animal models of plaque rupture. We previously reported that elastin fragmentation, due to a mutation (C1039G(+/-)) in the fibrillin-1 (Fbn1) gene, promotes atherogenesis and a highly unstable plaque phenotype in apolipoprotein E deficient (ApoE(-/-)) mice on a Western-type diet (WD). Here, we investigated whether plaque rupture occurred in ApoE(-/-)Fbn1(C1039G+/-) mice and was associated with myocardial infarction, stroke, and sudden death. METHODS AND RESULTS: Female ApoE(-/-)Fbn1(C1039G+/-) and ApoE(-/-) mice were fed a WD for up to 35 weeks. Compared to ApoE(-/-) mice, plaques of ApoE(-/-)Fbn1(C1039G+/-) mice showed a threefold increase in necrotic core size, augmented T-cell infiltration, a decreased collagen I content (70 ± 10%), extensive neovascularization, intraplaque haemorrhage, and a significant increase in matrix metalloproteinase-2, -9, -12, and -13 expression or activity. Plaque rupture was observed in 70% of ascending aortas and in 50% of brachiocephalic arteries of ApoE(-/-)Fbn1(C1039G+/-) mice. In ApoE(-/-) mice, plaque rupture was not seen in ascending aortas and only in 10% of brachiocephalic arteries. Seventy percent of ApoE(-/-)Fbn1(C1039G+/-) mice died suddenly, whereas all ApoE(-/-) mice survived. ApoE(-/-)Fbn1(C1039G+/-) mice showed coronary plaques and myocardial infarction (75% of mice). Furthermore, they displayed head tilt, disorientation, and motor disturbances (66% of cases), disturbed cerebral blood flow (73% of cases; MR angiograms) and brain hypoxia (64% of cases), indicative of stroke. CONCLUSIONS: Elastin fragmentation plays a key role in plaque destabilization and rupture. ApoE(-/-)Fbn1(C1039G+/-) mice represent a unique model of acute plaque rupture with human-like complications.


Assuntos
Morte Súbita/etiologia , Elastina/metabolismo , Infarto do Miocárdio/etiologia , Placa Aterosclerótica/etiologia , Acidente Vascular Cerebral/etiologia , Animais , Aorta , Apolipoproteínas E/deficiência , Biomarcadores/metabolismo , Tronco Braquiocefálico , Cardiomegalia/etiologia , Cardiomegalia/fisiopatologia , Artéria Carótida Primitiva , Circulação Cerebrovascular/fisiologia , Dieta Ocidental , Modelos Animais de Doenças , Feminino , Fibrilina-1 , Fibrilinas , Hemorragia/etiologia , Hipóxia Encefálica/etiologia , Hipóxia Encefálica/fisiopatologia , Camundongos , Proteínas dos Microfilamentos/deficiência , Microvasos , Infarto do Miocárdio/fisiopatologia , Neovascularização Patológica/etiologia , Neovascularização Patológica/fisiopatologia , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/fisiopatologia , Placa Aterosclerótica/fisiopatologia , Ruptura Espontânea/etiologia , Ruptura Espontânea/fisiopatologia , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/fisiopatologia
2.
J Vasc Res ; 50(6): 486-97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24192582

RESUMO

AIMS: Insulin resistance, dyslipidemia and hypertension are independent mediators of endothelial dysfunction. It is incompletely defined whether dyslipidemia and hypertension in addition to diabetes mellitus type II (DMII), as seen in the metabolic syndrome (MS), worsen diabetes-induced endothelial dysfunction. Furthermore, it is unclear whether treatment influences endothelial dysfunction similarly in MS and DMII. Therefore, we studied vascular reactivity and the effect of in vivo treatment with angiotensin-converting enzyme inhibition (ACE-I) or hypocaloric diet in LDL receptor- and leptin-deficient (ob/ob), double knockout mice (DKO), featuring MS and in ob/ob mice with DMII. METHODS AND RESULTS: Vascular reactivity was studied in isolated aortic ring segments. Maximum vasorelaxant response to acetylcholine (Ach) was more depressed in DKO than in ob/ob mice, whereas response to bradykinin (BK) was equally attenuated in both genotypes (52 ± 3 and 23 ± 9% reversal of preconstriction induced by 10(-7) M phenylephrine in DKO vs. 76 ± 3 and 23 ± 8% reversal of preconstriction in ob/ob mice, respectively). ACE-I and hypocaloric diet improved ACh-induced vasorelaxation significantly (89 ± 2 and 59 ± 2% reversal of preconstriction in DKO vs. 80 ± 3 and 84 ± 4% in ob/ob mice, respectively), but not the response to BK. CONCLUSION: These results indicate a differential impact of DMII and MS on endothelial function. ACE-I and hypocaloric diet improved ACh-, but not BK-induced vasorelaxation in these mouse models of DMII and MS.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Restrição Calórica , Captopril/farmacologia , Diabetes Mellitus Tipo 2/terapia , Dislipidemias/terapia , Endotélio Vascular/efeitos dos fármacos , Hipertensão/terapia , Óxido Nítrico/metabolismo , Redução de Peso , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Terapia Combinada , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Dislipidemias/sangue , Dislipidemias/genética , Dislipidemias/fisiopatologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Hipertensão/sangue , Hipertensão/genética , Hipertensão/fisiopatologia , Leptina/deficiência , Leptina/genética , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Tempo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
3.
Catheter Cardiovasc Interv ; 81(7): E259-68, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22887820

RESUMO

AIM: To evaluate differences in strut coverage, inflammation and endothelialization between two second-generation polymer-based drug-eluting stents (DES) in an atherosclerotic rabbit double-injury iliac artery model at 28 days follow-up. METHODS AND RESULTS: Rabbits with induced atheroma received bilateral iliac artery stents: everolimus-eluting stent (Xience V EES; Abbott Vascular), zotarolimus-eluting stent (Resolute ZES; Medtronic CardioVascular), or bare-metal stent (BMS; MultiLink Vision; Abbott Vascular). After 28 days, total neointimal coverage examined by scanning electron microscopy was >98% for all three stent types. Neointimal thickness above stent struts was decreased by 50% in Xience V EES (0.06 ± 0.01 mm; P = 0.00001) compared with BMS (0.15 ± 0.03 mm) and Resolute ZES (0.12 ± 0.04 mm). Luminal area was largest for Xience V EES (3.79 ± 0.33 mm(2) ; P = 0.0003 for Xience V EES vs. BMS), followed by Resolute ZES (3.46 ± 0.45 mm(2) ; P = 0.083 for Resolute ZES vs. BMS) and BMS (3.07 ± 0.53 mm(2) ). Percentage area stenosis was smallest for Xience V EES (17.23 ± 3.64%; P = 0.00001), while BMS (30.25 ± 7.48%) and Resolute ZES (30.79 ± 7.15%) did not differ. Endothelial monolayer regrowth was significantly lower in Resolute ZES (65 ± 13%) versus BMS (79 ± 11%; P = 0.004). There was no difference between Xience V EES (74 ± 10%) and BMS. Xience V EES was further associated with a lower number of inflammatory cells surrounding the stent struts (7 ± 2 per strut) in comparison to Resolute ZES (15 ± 6; P = 0.0001) and BMS (17 ± 9; P = 0.0005). CONCLUSION: In this atherosclerotic rabbit model, Xience V EES suppressed neointimal thickening better, with normal endothelial regrowth as compared with BMS, and less strut-induced inflammation.


Assuntos
Angioplastia com Balão/instrumentação , Aterosclerose/terapia , Stents Farmacológicos , Artéria Ilíaca/lesões , Lesões do Sistema Vascular/terapia , Angioplastia com Balão/efeitos adversos , Animais , Aterosclerose/diagnóstico por imagem , Aterosclerose/patologia , Fármacos Cardiovasculares/administração & dosagem , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/ultraestrutura , Everolimo , Artéria Ilíaca/diagnóstico por imagem , Artéria Ilíaca/ultraestrutura , Inflamação/etiologia , Inflamação/patologia , Inflamação/prevenção & controle , Masculino , Neointima , Desenho de Prótese , Coelhos , Radiografia , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Fatores de Tempo , Lesões do Sistema Vascular/diagnóstico por imagem , Lesões do Sistema Vascular/patologia
4.
Arterioscler Thromb Vasc Biol ; 32(5): 1228-35, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22345170

RESUMO

OBJECTIVE: Stent-based delivery of the mammalian target of rapamycin (mTOR) inhibitor everolimus is a promising strategy for the treatment of coronary artery disease. We studied potential adverse effects associated with mTOR inhibition. METHODS AND RESULTS: Macrophages in culture were either treated with everolimus or starved to inhibit mTOR. Everolimus led to inhibition of protein translation, activation of p38 MAPK, and the release of proinflammatory cytokines (eg, IL-6, TNFα) and chemokines (eg, MCP1, Rantes) before induction of autophagic death. These effects were also observed with rapamycin, but not after starvation. Everolimus-induced cytokine release was similar in macrophages lacking the essential autophagy gene Atg7 but was inhibited when macrophages were cotreated with p38 MAPK inhibitor SB202190 or the glucocorticoid clobetasol. Combined stent-based delivery of clobetasol and everolimus in rabbit plaques downregulated TNFα expression as compared with everolimus-treated plaques but did not affect the ability of everolimus to induce macrophage clearance. CONCLUSIONS: mTOR inhibition by everolimus triggers cytokine release in macrophages through inhibition of protein translation and p38 activation. These findings provide a rationale for combined local treatment of atherosclerotic plaques with everolimus and an anti-inflammatory agent.


Assuntos
Doença da Artéria Coronariana/cirurgia , Citocinas/biossíntese , Stents Farmacológicos , Glucocorticoides/farmacologia , Macrófagos/metabolismo , Sirolimo/análogos & derivados , Animais , Autofagia/efeitos dos fármacos , Western Blotting , Sobrevivência Celular , Células Cultivadas , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Everolimo , Humanos , Imuno-Histoquímica , Macrófagos/efeitos dos fármacos , Macrófagos/ultraestrutura , Camundongos , Microscopia Eletrônica de Transmissão , Desenho de Prótese , Coelhos , Sirolimo/farmacologia
5.
J Cardiovasc Pharmacol ; 61(4): 318-23, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23249678

RESUMO

In the mouse aorta, contractions evoked by the α(1)-adrenoceptor agonist phenylephrine are strongly suppressed by the continuous production of nitric oxide (NO). We investigated whether phenylephrine itself stimulated NO production by activating endothelial α(2)-adrenoceptors. On a prostaglandin F(2α) contraction, the α(2)-adrenoceptor agonist 5-bromo-N-(4,5-dihydro-1H-imidazol-2-yl)-6-quinoxalinamine (UK14304) induced 29.3 ± 7.4% relaxation, which was inhibited by 0.1 µM 2-[(4,5-Dihydro-1H-imidazol-2-yl)methyl]-2,3-dihydro-1-methyl-1H-isoindole (BRL44408) with a pKB' corresponding to α(2)-antagonism. In the presence of NO synthase blockers, UK14304 elicited small contractions above 1 µM that were inhibited by 0.1 µM prazosin, but not influenced by 0.1 µM rauwolscine. At 3 µM or higher concentrations, phenylephrine caused only modest relaxation (up to 7.4 ± 2.3%) of segments constricted with prostaglandin F(2α) in the presence of prazosin, which was abolished with 0.1 µM BRL44408. Furthermore, BRL44408 did not increase contractions induced with 1 µM phenylephrine. These results confirm that α(1)- but not α(2)-adrenoceptors are expressed on aortic smooth muscle cells, whereas endothelial cells only express α(2)-adrenoceptors. Moreover, phenylephrine exerted a very modest relaxing effect through nonspecific stimulation of α(2)-adrenoceptors, but only at concentrations higher than 1 µM. It is concluded that the high basal output of NO in the isolated mouse aorta is not due to stimulation of α-adrenoceptors.


Assuntos
Aorta Torácica/efeitos dos fármacos , Óxido Nítrico/metabolismo , Fenilefrina/farmacologia , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Aorta Torácica/metabolismo , Tartarato de Brimonidina , Relação Dose-Resposta a Droga , Imidazóis/administração & dosagem , Imidazóis/farmacologia , Isoindóis/administração & dosagem , Isoindóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase/metabolismo , Fenilefrina/administração & dosagem , Prazosina/farmacologia , Quinoxalinas/administração & dosagem , Quinoxalinas/farmacologia , Receptores Adrenérgicos alfa 2/metabolismo , Vasoconstritores/administração & dosagem , Vasoconstritores/farmacologia , Ioimbina/farmacologia
6.
Basic Res Cardiol ; 107(3): 269, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22543675

RESUMO

Atherosclerotic plaques tend to rupture as a consequence of a weakened fibrous cap, particularly in the shoulder regions where most macrophages reside. Macrophages express Toll-like receptors to recognize pathogens and eliminate intracellular pathogens by inducing autophagy. Because Toll-like receptor 7 (TLR7) is thought to be expressed in macrophages but not in smooth muscle cells (SMCs), we investigated whether induction of macrophage autophagic death by TLR7 ligand imiquimod can affect the composition of atherosclerotic plaques in favor of their stability. Immunohistochemical staining of human carotid plaques as well as Western blotting of cultured macrophages and SMCs confirmed that TLR7 was expressed in macrophages, but not in SMCs. In vitro experiments showed that only TLR7 expressing cells underwent imiquimod-induced cell death, which was characterized by autophagosome formation. Imiquimod-treated macrophages activated nuclear factor-κB (NF-κB) and released pro-inflammatory cytokines and chemokines. This effect was inhibited by the glucocorticoid dexamethasone. Imiquimod-induced cytokine release was significantly decreased in autophagy-deficient macrophages because these cells died by necrosis at an accelerated pace. Local in vivo administration of imiquimod to established atherosclerotic lesions in rabbit carotid arteries induced macrophage autophagy without induction of cell death, and triggered cytokine production, upregulation of vascular adhesion molecule-1, infiltration of T-lymphocytes, accumulation of macrophages and enlargement of plaque area. Treatment with dexamethasone suppressed these pro-inflammatory effects in vivo. SMCs and endothelial cells in imiquimod-treated plaques were not affected. In conclusion, imiquimod induces macrophage autophagy in atherosclerotic plaques, but stimulates plaque progression through cytokine release and enhanced infiltration of inflammatory cells.


Assuntos
Aminoquinolinas/farmacologia , Autofagia/efeitos dos fármacos , Inflamação/induzido quimicamente , Macrófagos/efeitos dos fármacos , Glicoproteínas de Membrana/agonistas , Placa Aterosclerótica/tratamento farmacológico , Receptor 7 Toll-Like/agonistas , Aminoquinolinas/toxicidade , Animais , Anti-Inflamatórios/farmacologia , Western Blotting , Citocinas/metabolismo , Dexametasona/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Imiquimode , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Necrose , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/patologia , Coelhos , Fatores de Tempo , Receptor 7 Toll-Like/deficiência , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Células U937
7.
Arterioscler Thromb Vasc Biol ; 31(11): 2552-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21799179

RESUMO

OBJECTIVE: The goal of this study was to examine the functional relationship between aging endothelium and thrombogenicity in a mouse model of premature aging. METHODS AND RESULTS: Coagulation tests and factors, blood cell counts, aorta endothelial function, aorta gene expression, and FeCl(3)-induced thrombosis in mesenteric blood vessels were analyzed in 10- to 30-week-old brain and muscle ARNT-like protein-1 (Bmal1)-deficient (knockout [KO]) mice and wild-type littermates. Ten-week-old KO mice manifested shortened prothrombin times (9.7 versus 11.3 seconds in wild-type) and elevated plasma fibrinogen (264 versus 172 mg/dL). At 30 weeks, factor VII (198% versus 149%), and platelet counts (2049 versus 1354 K/µL) were increased in KO mice. Gene deficiency reduced the vasoactive nitric oxide production at 10 and 30 weeks and tended to reduce and increase the protein expression of thrombomodulin and von Willebrand factor, respectively, with aging. Shortened venular and arteriolar occlusion times on FeCl(3)-induced injury in 10-week-old KO mice confirmed higher thrombogenicity, culminating in priapism, observed in 60% of 25- to 30-week-old KO males. CONCLUSION: Endothelial dysfunction and a hypercoagulable state cause early arterial and venous thrombogenicity in Bmal1 KO mice. With aging, progressive endothelial dysfunction, rising platelet counts, and high factor VII further enhance thrombogenicity, provoking priapism.


Assuntos
Fatores de Transcrição ARNTL/deficiência , Senilidade Prematura/metabolismo , Senilidade Prematura/fisiopatologia , Progressão da Doença , Trombose/metabolismo , Trombose/fisiopatologia , Fatores de Transcrição ARNTL/genética , Envelhecimento/fisiologia , Animais , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Fibrinogênio/metabolismo , Masculino , Camundongos , Camundongos Knockout , Contagem de Plaquetas , Tempo de Protrombina , Trombomodulina/metabolismo , Fator de von Willebrand/metabolismo
8.
BMC Physiol ; 12: 9, 2012 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-22943445

RESUMO

BACKGROUND: Electrophysiological studies of L-type Ca2+ channels in isolated vascular smooth muscle cells revealed that depolarization of these cells evoked a transient and a time-independent Ca2+ current. The sustained, non-inactivating current occurred at voltages where voltage-dependent activation and inactivation overlapped (voltage window) and its contribution to basal tone or active tension in larger multicellular blood vessel preparations is unknown at present. This study investigated whether window Ca2+ influx affects isometric contraction of multicellular C57Bl6 mouse aortic segments. RESULTS: Intracellular Ca2+ (Cai2+, Fura-2), membrane potential and isometric force were measured in aortic segments, which were clamped at fixed membrane potentials by increasing extracellular K+ concentrations. K+ above 20 mM evoked biphasic contractions, which were not affected by inhibition of IP3- or Ca2+ induced Ca2+ release with 2-aminoethoxydiphenyl borate or ryanodine, respectively, ruling out the contribution of intracellular Ca2+ release. The fast force component paralleled Cai2+ increase, but the slow contraction coincided with Cai2+ decrease. In the absence of extracellular Ca2+, basal tension and Cai2+ declined, and depolarization failed to evoke Cai2+ signals or contraction. Subsequent re-introduction of external Ca2+ elicited only slow contractions, which were now matched by Cai2+ increase. After Cai2+ attained steady-state, isometric force kept increasing due to Ca2+- sensitization of the contractile elements. The slow force responses displayed a bell-shaped voltage-dependence, were suppressed by hyperpolarization with levcromakalim, and enhanced by an agonist of L-type Ca2+ channels (BAY K8644). CONCLUSION: The isometric response of mouse aortic segments to depolarization consists of a fast, transient contraction paralleled by a transient Ca2+ influx via Ca2+ channels which completely inactivate. Ca2+ channels, which did not completely inactivate during the depolarization, initiated a second, sustained phase of contraction, which was matched by a sustained non-inactivating window Ca2+ influx. Together with sensitization, this window L-type Ca2+ influx is a major determinant of basal and active tension of mouse aortic smooth muscle.


Assuntos
Aorta/fisiologia , Cálcio/metabolismo , Contração Muscular/fisiologia , Músculo Liso Vascular/fisiologia , Animais , Aorta/metabolismo , Canais de Cálcio Tipo L/metabolismo , Fura-2/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Potássio/metabolismo
9.
Parasitology ; 139(11): 1429-37, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22776404

RESUMO

Leishmania parasites are able to survive in the macrophage, one of the most hostile environments of the vertebrate host. The present study investigated how Leishmania infection influences these host cell defence mechanisms. Macrophages were infected with antimony-susceptible and -resistant Leishmania strains. Free radical production in Leishmania-infected macrophages was measured by electron paramagnetic resonance. Apoptosis was detected with fluorescence microscopy using Annexin-V FITC labelling and with Western blotting to detect caspase-3 cleavage. Independent of their drug susceptibility profile or species background, all studied Leishmania strains induced a similar increase in free radical production in macrophages. O2 ●- production was significantly elevated during phagocytosis of the stationary phase promastigotes. Conversely, NO levels increased later in the infection and none of the strains induced capsase-3 cleavage. Leishmania donovani infection led to phosphatidylserine externalization only in RAW 264.7 cells. After an initial burst of O2 ●- during phagocytosis of promastigotes, amastigotes protect themselves by decreasing the O2 ●- production to the basal level. An increased NO production was observed 6 h after infection. Finally, induction of cell death is probably not essential in the survival of the parasite within the macrophage.


Assuntos
Apoptose/imunologia , Leishmaniose/imunologia , Macrófagos/imunologia , Macrófagos/parasitologia , Estresse Oxidativo/imunologia , Animais , Linhagem Celular Tumoral , Macrófagos/citologia , Camundongos , Óxido Nítrico/metabolismo , Superóxidos/metabolismo
10.
J Vasc Res ; 48(1): 31-42, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20606469

RESUMO

BACKGROUND/AIMS: Site-specific atherosclerosis is generally attributed to differential gene expression in endothelial cells. We investigated whether the transcriptome of smooth muscle cells is different between atherosclerosis-prone and atherosclerosis-resistant regions in apolipoprotein E-deficient (apoE-/-) mice before plaque development, and in C57Bl/6 mice. METHODS: De-endothelialized aortas (both strains: 3 males, 3 females, age 4 months) were divided into atherosclerosis-prone (AA: ascending aorta, aortic arch and proximal 2 mm of thoracic aorta) and -resistant (CTA: central thoracic aorta, i.e. 6 mm distal from the proximal 2 mm) regions. The transcriptome of these two regions was compared using whole-genome mouse microarrays. RESULTS: Microarray analysis revealed differential expression (>2-fold difference) of 70 and 244 genes in C57Bl/6 and apoE-/- mice. This was confirmed for 6 genes using the real-time quantitative polymerase chain reaction. Up- or downregulation in the AA was observed for 33 and 37 genes in C57Bl/6, and for 186 and 58 genes in apoE-/- mice, respectively. The 201 genes that showed exclusively differential expression in apoE-/- mice were related to atherosclerotic processes, such as cell adhesion, proliferation, differentiation, motility, cell death, lipid metabolism and immune responses. CONCLUSION: Our findings indicate that smooth muscle cells display an altered transcriptome at atherosclerosis-prone locations before actual lesion development.


Assuntos
Apolipoproteínas E/genética , Aterosclerose , Perfilação da Expressão Gênica , Músculo Liso Vascular/fisiologia , Fatores Etários , Animais , Aorta/patologia , Aorta/fisiopatologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Feminino , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Hipercolesterolemia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Liso Vascular/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica/fisiologia
11.
Clin Sci (Lond) ; 120(9): 415-26, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21143200

RESUMO

We investigated whether activation of circulating DCs (dendritic cells) or levels of Flt3L (FMS-like tyrosine kinase 3 ligand) and GM-CSF (granulocyte/macrophage colony-stimulating factor), haematopoietic growth factors important for DC differentiation, could account for reduced blood DC numbers in CAD (coronary artery disease) patients. Concentrations of Flt3L and GM-CSF were measured in plasma from CAD patients (n = 15) and controls (n = 12). Frequency and phenotype of mDCs (myeloid dendritic cells) and pDCs (plasmacytoid dendritic cells) were analysed by multicolour flow cytometry in fresh blood, and after overnight incubation with TLR (Toll-like receptor)-4 or -7 ligands LPS (lipopolysaccharide) or IQ (imiquimod). DC function was measured by IL (interleukin)-12 and IFN (interferon)-α secretion. Circulating numbers of CD11c+ mDCs and CD123+ pDCs and frequencies of CD86+ and CCR-7+ (CC chemokine receptor type 7) mDCs, but not pDCs, were declined in CAD. In addition, plasma Flt3L, but not GM-CSF, was lower in patients and positively correlated with blood DC counts. In response to LPS, mDCs up-regulated CD83 and CD86, but CCR-7 expression and IL-12 secretion remained unchanged, similarly in patients and controls. Conversely, pDCs from patients had lower CD83 and CCR-7 expression after overnight incubation and had a weaker IQ-induced up-regulation of CD83 and IFN-α secretion. In conclusion, our results suggest that reduced blood DC counts in CAD are, at least partly, due to impaired DC differentiation from bone marrow progenitors. Decreased levels of mDCs are presumably also explained by activation and subsequent migration to atherosclerotic plaques or lymph nodes. Although mDCs are functioning normally, pDCs from patients appeared to be both numerically and functionally impaired.


Assuntos
Doença da Artéria Coronariana/sangue , Células Dendríticas/metabolismo , Proteínas de Membrana/sangue , Estudos de Casos e Controles , Contagem de Células , Diferenciação Celular/fisiologia , Células Cultivadas , Células Dendríticas/fisiologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/sangue , Humanos , Imunofenotipagem , Interferon-alfa/sangue , Interleucina-12/sangue , Masculino , Pessoa de Meia-Idade
12.
Arterioscler Thromb Vasc Biol ; 30(7): 1415-22, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20413730

RESUMO

OBJECTIVE: Apoptosis is a key feature of advanced atherosclerotic plaques. Attraction signals such as p43 released from apoptotic cells play a crucial role in the timely removal of the apoptotic remnants by recruiting fresh phagocytes. Here, we sought to determine whether p43 may link apoptosis to inflammation and plaque progression. METHODS AND RESULTS: RT-PCR and immunohistochemistry showed that p43 was abundantly expressed in human plaques compared with nonatherosclerotic mammary arteries and colocalized with splicing factor SC-35. Cell culture experiments indicated that p43 expression was associated with enhanced protein translation. On initiation of apoptosis or necrosis, p43 was cleaved by calpains and released as truncated protein p43(apoptosis-released factor [ARF]). Processing of p43 into endothelial monocyte activating polypeptide II was not observed. Full-length p43, but not p43(ARF) or endothelial monocyte activating polypeptide II, activated THP1 monocytes (upregulation of tumor necrosis factor alpha, interleukin 1 beta, interleukin 8, macrophage inflammatory protein (MIP)-1 alpha, MIP1 beta, MIP2 alpha) and endothelial cells (enhanced synthesis of E-selectin, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, tissue factor). The chemotactic activity of p43 or fragments thereof was poor compared with ATP. Treatment of smooth muscle cells with p43 did not induce cell death. CONCLUSIONS: p43 is cleaved during apoptosis by calpains and released as a truncated protein that is harmless for the structure of the plaque.


Assuntos
Apoptose , Aterosclerose/metabolismo , Doenças das Artérias Carótidas/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo , Proteínas de Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas de Ligação a RNA/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Calpaína/metabolismo , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/imunologia , Doenças das Artérias Carótidas/patologia , Estudos de Casos e Controles , Células Cultivadas , Doença da Artéria Coronariana/sangue , Citocinas/sangue , Citocinas/genética , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas de Neoplasias/sangue , Proteínas de Neoplasias/genética , Proteínas Nucleares/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/sangue , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteínas/metabolismo , Fatores de Processamento de Serina-Arginina , Fatores de Tempo , Transfecção , Regulação para Cima
13.
Mediators Inflamm ; 2011: 941396, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21976788

RESUMO

BACKGROUND: Atherosclerosis is a chronic inflammatory disease with atherosclerotic plaques containing inflammatory infiltrates predominantly consisting of monocytes/macrophages and activated T cells. More recent is the implication of dendritic cells (DCs) in the disease. Since DCs were demonstrated in human arteries in 1995, numerous studies in humans suggest a role for these professional antigen-presenting cells in atherosclerosis. AIM: This paper focuses on the observations made in blood and arteries of patients with atherosclerosis. In principal, flow cytometric analyses show that circulating myeloid (m) and plasmacytoid (p) DCs are diminished in coronary artery disease, while immunohistochemical studies describe increased intimal DC counts with evolving plaque stages. Moreover, mDCs and pDCs appear to behave differently in atherosclerosis. Yet, the origin of plaque DCs and their relationship with blood DCs are unknown. Therefore, several explanations for the observed changes are postulated. In addition, the technical challenges and discrepancies in the research field are discussed. FUTURE: Future studies in humans, in combination with experimental animal studies will unravel mechanisms leading to altered blood and plaque DCs in atherosclerosis. As DCs are crucial for inducing but also dampening immune responses, understanding their life cycle, trafficking and function in atherosclerosis will determine potential use of DCs in antiatherogenic therapies.


Assuntos
Aterosclerose/imunologia , Células Dendríticas/imunologia , Placa Aterosclerótica/imunologia , Animais , Células Dendríticas/citologia , Citometria de Fluxo , Humanos , Placa Aterosclerótica/metabolismo
14.
Circulation ; 120(24): 2478-87, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19948973

RESUMO

BACKGROUND: Arterial stiffness has been associated with an increased cardiovascular risk. The aim of this study was to investigate the interaction between arterial stiffness and atherosclerosis. METHODS AND RESULTS: Mice with a mutation C1039G+/-) in the fibrillin-1 gene leading to fragmentation of the elastic fibers were crossbred with apolipoprotein E-deficient (ApoE-/-) mice. Subsequently, ApoE-/- and ApoE-/-C1039G+/- mice were fed a Western-type diet for 10 or 20 weeks. Our results show that the interaction between arterial stiffness and atherosclerosis is bidirectional. On the one hand, arterial stiffness in ApoE-/-C1039G+/- mice increased more rapidly in the presence of atherosclerotic plaques. On the other hand, arterial stiffness promoted the development of larger and more unstable plaques in ApoE-/-C1039G+/- mice. The plaque area at the aortic root was increased 1.5- and 2.1-fold in ApoE-/-C1039G+/- mice after 10 and 20 weeks of Western-type diet, respectively. After 10 weeks of Western-type diet, plaques of ApoE-/-C1039G+/- mice showed increased apoptosis of smooth muscle cells, which was associated with a decrease in collagen content, an enlargement of the necrotic core, and an increase in macrophages. After 20 weeks of Western-type diet, the number of buried fibrous caps was increased in atherosclerotic lesions of ApoE-/-C1039G+/- mice, not only at the level of the aortic valves but also in the brachiocephalic artery and in the upper, middle, and lower thoracic aorta. Furthermore, acute plaque rupture was observed. CONCLUSIONS: These results indicate that fragmentation of the elastic fibers leads to increased vascular stiffness, which promotes features of multifocal plaque instability.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/genética , Aterosclerose/fisiopatologia , Proteínas dos Microfilamentos/fisiologia , Animais , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatologia , Apolipoproteínas E/genética , Aterosclerose/metabolismo , Feminino , Fibrilina-1 , Fibrilinas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética
15.
Basic Res Cardiol ; 105(1): 39-50, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19693627

RESUMO

The ubiquitin-proteasome system is involved in the development and progression of atherosclerosis. The aim of this study was to investigate whether plaque composition is affected by proteasome function. In vitro, the potent and selective proteasome inhibitor bortezomib induced apoptosis in both cultured smooth muscle cells (SMCs) and activated macrophages. This effect was associated with increased expression of C/EBP homologous protein and cleavage of caspase-12, indicative of endoplasmic reticulum stress. The sensitivity to the proapoptotic effects of proteasome inhibition correlated with the protein synthesis rate. Proteasome inhibition in explanted atherosclerotic plaques of ApoE-deficient mice resulted in a significant decrease in SMCs and macrophages, indicating that both cell types in the atherosclerotic plaque were susceptible to the proapoptotic effects of proteasome inhibition. In vivo proteasome inhibition in ApoE-deficient mice did not affect plaque size or composition of early atherosclerotic plaques, but resulted in a significant decrease in collagen content as well as a significant enlargement of the necrotic core in advanced atherosclerotic plaques. In conclusion, our results indicate that an impaired proteasome function promotes features of a more rupture-prone plaque phenotype.


Assuntos
Aterosclerose/metabolismo , Ácidos Borônicos/farmacologia , Macrófagos/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirazinas/farmacologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apoptose/efeitos dos fármacos , Bortezomib , Células Cultivadas , Feminino , Camundongos , Camundongos Knockout , Fenótipo , Inibidores de Proteassoma , Coelhos , Ruptura Espontânea
16.
Basic Res Cardiol ; 105(1): 51-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19693628

RESUMO

Rupture-prone and ruptured plaques are characterized by the presence of large numbers of macrophages. N1177 is a contrast agent consisting of iodinated nanoparticles that are selectively phagocytosed by macrophages. The aim of this study was to investigate the effect of N1177 on the CT attenuation of rupture-prone and ruptured plaques in rabbits. In addition, we examined in vitro whether uptake of N1177 occurred without cytotoxic or pro-inflammatory effects on macrophages. In vitro, the viability of J774 macrophages was not affected by treatment with N1177. Moreover, N1177 had no effect on the phagocytic capacity or cytokine production of macrophages. For the in vivo experiments, 6 New Zealand White rabbits were fed a cholesterol-supplemented diet for 12-15 months, resulting in the development of large atherosclerotic plaques that resembled rupture-prone plaques in humans. In three rabbits, mechanical plaque rupture was induced by retrograde pullback of an embolic protection device. N1177 had no effect on the median density of rupture-prone plaques [35 HU (range 3-85) before injection vs. 32 HU (range 1-93) 2 h after injection of N1177; P > 0.05]. However, after induction of mechanical plaque rupture, the median density of the atherosclerotic plaques increased from 40 HU (range 6-86) before injection to 74 HU (range 14-111) 2 h after injection of N1177 (P < 0.001). Using time-of-flight static secondary ion mass spectrometry, the presence of N1177 nanoparticles was demonstrated in macrophage-rich areas of ruptured plaques, but not of non-ruptured plaques. In conclusion, our results show that N1177 is a contrast agent that can identify ruptured atherosclerotic plaques.


Assuntos
Aterosclerose/diagnóstico por imagem , Meios de Contraste , Iodo , Macrófagos/efeitos dos fármacos , Nanopartículas , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Contraste/efeitos adversos , Camundongos , Coelhos , Ruptura Espontânea/diagnóstico por imagem , Tomografia Computadorizada por Raios X
17.
J Vasc Res ; 47(3): 231-40, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19893320

RESUMO

AIM: Transglutaminase 2 (TG2) is important for the deposition and stability of the extracellular matrix via effects on cross-linking of matrix proteins and transforming growth factor beta (TGFbeta) activity. The purpose of this study was to investigate the effect of TG2 deficiency on the composi- tion of atherosclerotic plaques. METHODS: Apolipoprotein E (ApoE)(-/-) mice were crossbred with TG2(-/-) mice to obtain ApoE(-/-)TG2(-/-) mice. ApoE(-/-) and ApoE(-/-)TG2(-/-) mice were fed a Western-type diet for 16 or 30 weeks to determine the effect of TG2 deficiency on early and advanced atherosclerosis, respectively. RESULTS: Atherosclerotic plaques of ApoE(-/-)TG2(-/-) mice showed decreased cross-linking of matrix proteins, as well as decreased nuclear staining for phospho-Smad2/-Smad3, indicative of decreased TGFbeta activity. Compared to ApoE(-/-) mice, plaque area was decreased by 45 and 48% in ApoE(-/-)TG2(-/-) mice after 16 and 30 weeks, respectively. Sirius red staining showed a significant decrease in collagen content in early and advanced atherosclerotic plaques of ApoE(-/-)TG2(-/-) mice. Furthermore, there was a significant increase in macrophages in advanced atherosclerotic plaques of ApoE(-/-)TG2(-/-) mice. CONCLUSION: TG2 deficiency resulted in a decreased collagen content and increased inflammation, which are features of a more unstable plaque.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/enzimologia , Proteínas de Ligação ao GTP/deficiência , Inflamação/enzimologia , Transglutaminases/deficiência , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Proteínas da Matriz Extracelular/metabolismo , Feminino , Fibrose , Proteínas de Ligação ao GTP/genética , Inflamação/genética , Inflamação/patologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Glutamina gama-Glutamiltransferase , Ruptura , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Transglutaminases/genética
18.
J Cardiovasc Pharmacol ; 55(3): 269-75, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20051876

RESUMO

Because macrophages play an important role in the destabilization of atherosclerotic plaques, and smooth muscle cells (SMCs) contribute to plaque stability, selective clearance of macrophages in atherosclerotic plaques is a promising strategy for plaque stabilization. In the present study, we investigated the effects of fluvastatin, simvastatin, lovastatin, and pravastatin on the viability of macrophages and SMCs. All statins, except pravastatin, induced cell death of J774A.1 macrophages after 24 hours, albeit with different sensitivity. The viability of rabbit aortic SMCs was hardly affected. Fluvastatin-induced macrophage cell death was characterized as apoptosis and could be reversed by isoprenoid intermediates of the mevalonate pathway. Peritoneal macrophages from male or female mice were much more resistant to statin-induced cell death. The high sensitivity of J774A.1 macrophages to statin-induced cell death was related to the strong 3-hydroxy-3-methylglutaryl coenzyme A reductase activity in these cells. Macrophage and SMC content of rabbit atherosclerotic plaques was not affected after in vitro treatment with fluvastatin or lovastatin for 3 days. In conclusion, fluvastatin, simvastatin, and lovastatin, but not pravastatin, can selectively induce apoptosis in J774A.1 macrophages, but not in SMCs, primary macrophages or rabbit atherosclerotic plaques. This effect was related to the degree of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity in the different cell types.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Macrófagos/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Feminino , Hidroximetilglutaril-CoA Redutases/efeitos dos fármacos , Hidroximetilglutaril-CoA Redutases/metabolismo , Macrófagos/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Coelhos
19.
Arterioscler Thromb Vasc Biol ; 28(3): 511-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18218989

RESUMO

OBJECTIVE: Recent studies proposed a pathogenic role for C-reactive protein (CRP), an independent predictor of cardiovascular disease (CVD), in atherosclerosis. Therefore, we tested whether CRP may modulate dendritic cell (DC) function, because these professional antigen-presenting cells have been implicated in atherogenesis. METHODS AND RESULTS: Human monocyte-derived immature DCs were cultured with human CRP (0 to 60 microg/mL) for 24 hours. Thereafter, activation markers were measured by flow-cytometry and DCs were cocultured with CFSE-labeled lymphocytes to measure T-cell proliferation and interferon (IFN)-gamma secretion after 8 days. Exposure to 60 microg/mL CRP (n=5) induced an activated cell morphology and significant (CD40 increase MFI 5.23+/-0.28, P<0.01 paired t test; CD80 6.18+/-0.51, P<0.01) to modest (CD83 1.38+/-0.17, P<0.05, CCR7 1.60+/-0.29, P=0.05) upregulation of DC activation markers. The expression of CD86 and HLA-DR was high, but not affected. T-lymphocytes incubated with CRP-pulsed DCs displayed increased IFN-gamma secretion and proliferation (P<0.001). DC activation was concentration-dependent and detected from 2 mug/mL CRP; the maximum effect was equivalent to that seen with 0.1 microg/mL lipopolysaccharide (LPS). Polymyxin B abolished the LPS response, without influencing CRP effects. Finally, immunohistochemistry could demonstrate DC/CRP colocalization in human atherosclerotic lesions. CONCLUSIONS: These findings suggest that CRP in plaques or found circulating in CVD patients can influence DC function during atherogenesis.


Assuntos
Aterosclerose/metabolismo , Proteína C-Reativa/metabolismo , Células Dendríticas/metabolismo , Ativação Linfocitária/fisiologia , Análise de Variância , Proliferação de Células , Células Cultivadas , Células Dendríticas/citologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Antígenos HLA-DR/metabolismo , Humanos , Interferon gama/metabolismo , Monócitos/citologia , Polimixinas/farmacologia , Polissacarídeos/farmacologia , Probabilidade , Sensibilidade e Especificidade , Linfócitos T/citologia
20.
Mol Pharmacol ; 74(3): 777-84, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18523137

RESUMO

P2Y receptors are G-protein-coupled receptors activated by extracellular nucleotides. The P2Y(6) receptor is selectively activated by UDP, and its transcript has been detected in numerous organs, including the spleen, thymus, intestine, blood leukocytes, and aorta. To investigate the biological functions of this receptor, we generated P2Y(6)-null mice by gene targeting. The P2Y(6) knockout (KO) mice are viable and are not distinguishable from the wild-type (WT) mice in terms of growth or fertility. In thioglycollate-elicited macrophages, the production of inositol phosphate in response to UDP stimulation was lost, indicating that P2Y(6) is the unique UDP-responsive receptor expressed by mouse macrophages. Furthermore, the amount of interleukin-6 and macrophage-inflammatory protein-2, but not tumor necrosis factor-alpha, released in response to lipopolysaccharide stimulation was significantly enhanced in the presence of UDP, and this effect was lost in the P2Y(6) KO macrophages. The endothelium-dependent relaxation of the aorta by UDP was abolished in KO P2Y(6) mice. The contractile effect of UDP on the aorta, observed when endothelial nitric-oxide synthase is blocked, was also abolished in P2Y(6)-null mice. In conclusion, we generated P2Y(6)-deficient mice and have shown that these mice have a defective response to UDP in macrophages, endothelial cells, and vascular smooth muscle cells. These observations might be relevant to several physiopathological conditions such as atherosclerosis or hypertension.


Assuntos
Células Endoteliais/metabolismo , Macrófagos Peritoneais/metabolismo , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Citocinas/biossíntese , Células Endoteliais/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Fosfatos de Inositol/biossíntese , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/enzimologia , Camundongos , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Relaxamento Muscular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Nucleotídeos/farmacologia , Fenilefrina/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Purinérgicos P2/genética , Tioglicolatos/farmacologia , Sistema Vasomotor/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA