Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 4(11)2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31167964

RESUMO

Atherosclerotic plaques feature local proliferation of leukocytes and vascular smooth muscle cells (VSMCs) and changes in cellular metabolism. Yet the relationship between glucose utilization and proliferation has been technically impossible to study directly in cells of atherosclerotic plaques in vivo. We used multi-isotope imaging mass spectrometry (MIMS), a quantitative imaging platform, to measure coincident cell division and glucose utilization at suborganelle resolution in atherosclerotic plaques. In established plaques, 65% of intimal foam cells and only 4% of medial VSMCs were labeled with 15N-thymidine after 1 week of isotope treatment. Dividing cells demonstrated heightened glucose labeling. MIMS detected 2H-glucose label in multiple subcellular compartments within foam cells, including lipid droplets, the cytosol, and chromatin. Unexpectedly, we identified an intensely focal region of 2H-label in VSMCs underlying plaques. This signal diminished in regions of aorta without atherosclerosis. In advanced plaques, 15N-thymidine and 2H-glucose labeling in foam cells and VSMCs significantly decreased. These data demonstrate marked heterogeneity in VSMC glucose metabolism that was dependent on both proliferative status and proximity of VSMCs to plaques. Furthermore, these results reveal how quantitative mass spectrometry coupled with isotope imaging can complement other methods used to study cell biology directly in the growing atherosclerotic plaque in vivo.


Assuntos
Aterosclerose/diagnóstico por imagem , Proliferação de Células/fisiologia , Espectrometria de Massas/métodos , Imagem Molecular/métodos , Animais , Aorta/citologia , Aorta/diagnóstico por imagem , Aorta/metabolismo , Aterosclerose/metabolismo , Modelos Animais de Doenças , Células Espumosas/metabolismo , Leucócitos/citologia , Leucócitos/metabolismo , Masculino , Camundongos , Músculo Liso Vascular/diagnóstico por imagem , Músculo Liso Vascular/metabolismo , Isótopos de Nitrogênio/química , Isótopos de Nitrogênio/metabolismo , Timidina/química , Timidina/metabolismo
2.
Int J Mol Sci ; 20(11)2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-31159424

RESUMO

The PI3K/Akt pathway plays a crucial role in the survival, proliferation, and migration of macrophages, which may impact the development of atherosclerosis. Changes in Akt isoforms or modulation of the Akt activity levels in macrophages significantly affect their polarization phenotype and consequently atherosclerosis in mice. Moreover, the activity levels of Akt signaling determine the viability of monocytes/macrophages and their resistance to pro-apoptotic stimuli in atherosclerotic lesions. Therefore, elimination of pro-apoptotic factors as well as factors that antagonize or suppress Akt signaling in macrophages increases cell viability, protecting them from apoptosis, and this markedly accelerates atherosclerosis in mice. In contrast, inhibition of Akt signaling by the ablation of Rictor in myeloid cells, which disrupts mTORC2 assembly, significantly decreases the viability and proliferation of blood monocytes and macrophages with the suppression of atherosclerosis. In addition, monocytes and macrophages exhibit a threshold effect for Akt protein levels in their ability to survive. Ablation of two Akt isoforms, preserving only a single Akt isoform in myeloid cells, markedly compromises monocyte and macrophage viability, inducing monocytopenia and diminishing early atherosclerosis. These recent advances in our understanding of Akt signaling in macrophages in atherosclerosis may have significant relevance in the burgeoning field of cardio-oncology, where PI3K/Akt inhibitors being tested in cancer patients can have significant cardiovascular and metabolic ramifications.


Assuntos
Aterosclerose/etiologia , Aterosclerose/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Animais , Apoptose , Aterosclerose/patologia , Células Sanguíneas/metabolismo , Sobrevivência Celular , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Isoformas de Proteínas
3.
Arterioscler Thromb Vasc Biol ; 39(2): 156-169, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30567482

RESUMO

Objective- Macrophages express 3 Akt (protein kinase B) isoforms, Akt1, Akt2, and Akt3, which display isoform-specific functions but may be redundant in terms of Akt survival signaling. We hypothesize that loss of 2 Akt isoforms in macrophages will suppress their ability to survive and modulate the development of atherosclerosis. Approach and Results- To test this hypothesis, we reconstituted male Ldlr-/- mice with double Akt2/Akt3 knockout hematopoietic cells expressing only the Akt1 isoform (Akt1only). There were no differences in body weight and plasma lipid levels between the groups after 8 weeks of the Western diet; however, Akt1only→ Ldlr-/- mice developed smaller (57.6% reduction) atherosclerotic lesions with more apoptotic macrophages than control mice transplanted with WT (wild type) cells. Next, male and female Ldlr-/- mice were reconstituted with double Akt1/Akt2 knockout hematopoietic cells expressing the Akt3 isoform (Akt3only). Female and male Akt3only→ Ldlr-/- recipients had significantly smaller (61% and 41%, respectively) lesions than the control WT→ Ldlr-/- mice. Loss of 2 Akt isoforms in hematopoietic cells resulted in markedly diminished levels of white blood cells, B cells, and monocytes and compromised viability of monocytes and peritoneal macrophages compared with WT cells. In response to lipopolysaccharides, macrophages with a single Akt isoform expressed low levels of inflammatory cytokines; however, Akt1only macrophages were distinct in expressing high levels of antiapoptotic Il10 compared with WT and Akt3only cells. Conclusions- Loss of 2 Akt isoforms in hematopoietic cells, preserving only a single Akt1 or Akt3 isoform, markedly compromises monocyte and macrophage viability and diminishes early atherosclerosis in Ldlr-/- mice.


Assuntos
Aterosclerose/prevenção & controle , Macrófagos/fisiologia , Monócitos/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores de LDL/fisiologia , Animais , Sobrevivência Celular , Feminino , Sistema Hematopoético/citologia , Sistema Hematopoético/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/fisiologia
4.
Front Immunol ; 9: 215, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29487597

RESUMO

Background: Rictor is an essential component of mammalian target of rapamycin (mTOR) complex 2 (mTORC2), a conserved serine/threonine kinase that may play a role in cell proliferation, survival and innate or adaptive immune responses. Genetic loss of Rictor inactivates mTORC2, which directly activates Akt S473 phosphorylation and promotes pro-survival cell signaling and proliferation. Methods and results: To study the role of mTORC2 signaling in monocytes and macrophages, we generated mice with myeloid lineage-specific Rictor deletion (MRictor-/-). These MRictor-/- mice exhibited dramatic reductions of white blood cells, B-cells, T-cells, and monocytes but had similar levels of neutrophils compared to control Rictor flox-flox (Rictorfl/fl) mice. MRictor-/- bone marrow monocytes and peritoneal macrophages expressed reduced levels of mTORC2 signaling and decreased Akt S473 phosphorylation, and they displayed significantly less proliferation than control Rictorfl/fl cells. In addition, blood monocytes and peritoneal macrophages isolated from MRictor-/- mice were significantly more sensitive to pro-apoptotic stimuli. In response to LPS, MRictor-/- macrophages exhibited the M1 phenotype with higher levels of pro-inflammatory gene expression and lower levels of Il10 gene expression than control Rictorfl/fl cells. Further suppression of LPS-stimulated Akt signaling with a low dose of an Akt inhibitor, increased inflammatory gene expression in macrophages, but genetic inactivation of Raptor reversed this rise, indicating that mTORC1 mediates this increase of inflammatory gene expression. Next, to elucidate whether mTORC2 has an impact on atherosclerosis in vivo, female and male Ldlr null mice were reconstituted with bone marrow from MRictor-/- or Rictorfl/fl mice. After 10 weeks of the Western diet, there were no differences between the recipients of the same gender in body weight, blood glucose or plasma lipid levels. However, both female and male MRictor-/- → Ldlr-/- mice developed smaller atherosclerotic lesions in the distal and proximal aorta. These lesions contained less macrophage area and more apoptosis than lesions of control Rictorfl/fl → Ldlr-/- mice. Thus, loss of Rictor and, consequently, mTORC2 significantly compromised monocyte/macrophage survival, and this markedly diminished early atherosclerosis in Ldlr-/- mice. Conclusion: Our results demonstrate that mTORC2 is a key signaling regulator of macrophage survival and its depletion suppresses early atherosclerosis.


Assuntos
Aterosclerose/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Proteína Companheira de mTOR Insensível à Rapamicina/imunologia , Receptores de LDL/genética , Animais , Aorta/imunologia , Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Proliferação de Células , Sobrevivência Celular/imunologia , Dieta Ocidental/efeitos adversos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Companheira de mTOR Insensível à Rapamicina/genética
5.
Circ J ; 80(11): 2259-2268, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27725526

RESUMO

Macrophage apoptosis and the ability of macrophages to clean up dead cells, a process called efferocytosis, are crucial determinants of atherosclerosis lesion progression and plaque stability. Environmental stressors initiate endoplasmic reticulum (ER) stress and activate the unfolded protein response (UPR). Unresolved ER stress with activation of the UPR initiates apoptosis. Macrophages are resistant to apoptotic stimuli, because of activity of the PI3K/Akt pathway. Macrophages express 3 Akt isoforms, Akt1, Akt2 and Akt3, which are products of distinct but homologous genes. Akt displays isoform-specific effects on atherogenesis, which vary with different vascular cell types. Loss of macrophage Akt2 promotes the anti-inflammatory M2 phenotype and reduces atherosclerosis. However, Akt isoforms are redundant with regard to apoptosis. c-Jun NH2-terminal kinase (JNK) is a pro-apoptotic effector of the UPR, and the JNK1 isoform opposes anti-apoptotic Akt signaling. Loss of JNK1 in hematopoietic cells protects macrophages from apoptosis and accelerates early atherosclerosis. IκB kinase α (IKKα, a member of the serine/threonine protein kinase family) plays an important role in mTORC2-mediated Akt signaling in macrophages, and IKKα deficiency reduces macrophage survival and suppresses early atherosclerosis. Efferocytosis involves the interaction of receptors, bridging molecules, and apoptotic cell ligands. Scavenger receptor class B type I is a critical mediator of macrophage efferocytosis via the Src/PI3K/Rac1 pathway in atherosclerosis. Agonists that resolve inflammation offer promising therapeutic potential to promote efferocytosis and prevent atherosclerotic clinical events. (Circ J 2016; 80: 2259-2268).


Assuntos
Aterosclerose/metabolismo , Estresse do Retículo Endoplasmático , Macrófagos/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas , Animais , Aterosclerose/patologia , Humanos , Quinase I-kappa B/metabolismo , Isoenzimas/metabolismo , Macrófagos/patologia , Alvo Mecanístico do Complexo 2 de Rapamicina , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 36(6): 1122-31, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27102962

RESUMO

OBJECTIVE: The c-Jun NH2-terminal kinases (JNK) are regulated by a wide variety of cellular stresses and have been implicated in apoptotic signaling. Macrophages express 2 JNK isoforms, JNK1 and JNK2, which may have different effects on cell survival and atherosclerosis. APPROACH AND RESULTS: To dissect the effect of macrophage JNK1 and JNK2 on early atherosclerosis, Ldlr(-/-) mice were reconstituted with wild-type, Jnk1(-/-), and Jnk2(-/-) hematopoietic cells and fed a high cholesterol diet. Jnk1(-/-)→Ldlr(-/-) mice have larger atherosclerotic lesions with more macrophages and fewer apoptotic cells than mice transplanted with wild-type or Jnk2(-/-) cells. Moreover, genetic ablation of JNK to a single allele (Jnk1(+/-)/Jnk2(-/-) or Jnk1(-/-)/Jnk2(+/-)) in marrow of Ldlr(-/-) recipients further increased atherosclerosis compared with Jnk1(-/-)→Ldlr(-/-) and wild-type→Ldlr(-/-) mice. In mouse macrophages, anisomycin-mediated JNK signaling antagonized Akt activity, and loss of Jnk1 gene obliterated this effect. Similarly, pharmacological inhibition of JNK1, but not JNK2, markedly reduced the antagonizing effect of JNK on Akt activity. Prolonged JNK signaling in the setting of endoplasmic reticulum stress gradually extinguished Akt and Bad activity in wild-type cells with markedly less effects in Jnk1(-/-) macrophages, which were also more resistant to apoptosis. Consequently, anisomycin increased and JNK1 inhibitors suppressed endoplasmic reticulum stress-mediated apoptosis in macrophages. We also found that genetic and pharmacological inhibition of phosphatase and tensin homolog abolished the JNK-mediated effects on Akt activity, indicating that phosphatase and tensin homolog mediates crosstalk between these pathways. CONCLUSIONS: Loss of Jnk1, but not Jnk2, in macrophages protects them from apoptosis, increasing cell survival, and this accelerates early atherosclerosis.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Apoptose , Aterosclerose/enzimologia , Células da Medula Óssea/enzimologia , Macrófagos/enzimologia , Proteína Quinase 8 Ativada por Mitógeno/deficiência , Receptores de LDL/deficiência , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apoptose/efeitos dos fármacos , Aterosclerose/genética , Aterosclerose/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Sobrevivência Celular , Células Cultivadas , Dieta Hiperlipídica , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático , Predisposição Genética para Doença , Hipercolesterolemia/enzimologia , Hipercolesterolemia/genética , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 9 Ativada por Mitógeno/deficiência , Proteína Quinase 9 Ativada por Mitógeno/genética , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Placa Aterosclerótica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de LDL/genética , Transdução de Sinais , Proteína de Morte Celular Associada a bcl/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 36(4): 598-607, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26848161

RESUMO

OBJECTIVE: The IκB kinase (IKK) is an enzyme complex that initiates the nuclear factor κB transcription factor cascade, which is important in regulating multiple cellular responses. IKKα is directly associated with 2 major prosurvival pathways, PI3K/Akt and nuclear factor κB, but its role in cell survival is not clear. Macrophages play critical roles in the pathogenesis of atherosclerosis, yet the impact of IKKα signaling on macrophage survival and atherogenesis remains unclear. APPROACH AND RESULTS: Here, we demonstrate that genetic IKKα deficiency, as well as pharmacological inhibition of IKK, in mouse macrophages significantly reduces Akt S(473) phosphorylation, which is accompanied by suppression of mTOR complex 2 signaling. Moreover, IKKα null macrophages treated with lipotoxic palmitic acid exhibited early exhaustion of Akt signaling compared with wild-type cells. This was accompanied by a dramatic decrease in the resistance of IKKα(-/-) monocytes and macrophages to different proapoptotic stimuli compared with wild-type cells. In vivo, IKKα deficiency increased macrophage apoptosis in atherosclerotic lesions and decreased early atherosclerosis in both female and male low-density lipoprotein receptor (LDLR)(-/-) mice reconstituted with IKKα(-/-) hematopoietic cells and fed with the Western diet for 8 weeks compared with control LDLR(-/-) mice transplanted with wild-type cells. CONCLUSIONS: Hematopoietic IKKα deficiency in mouse suppresses Akt signaling, compromising monocyte/macrophage survival and this decreases early atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Quinase I-kappa B/deficiência , Macrófagos Peritoneais/enzimologia , Proteínas Proto-Oncogênicas c-akt , Animais , Apoptose , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Sobrevivência Celular , Células Cultivadas , Dieta Ocidental , Modelos Animais de Doenças , Feminino , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/genética , Mediadores da Inflamação/metabolismo , Fígado/embriologia , Fígado/enzimologia , Transplante de Fígado , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Masculino , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo
8.
J Lipid Res ; 56(8): 1449-60, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26059978

RESUMO

Macrophage apoptosis and efferocytosis are key determinants of atherosclerotic plaque inflammation and necrosis. Bone marrow transplantation studies in ApoE- and LDLR-deficient mice revealed that hematopoietic scavenger receptor class B type I (SR-BI) deficiency results in severely defective efferocytosis in mouse atherosclerotic lesions, resulting in a 17-fold higher ratio of free to macrophage-associated dead cells in lesions containing SR-BI(-/-) cells, 5-fold more necrosis, 65.2% less lesional collagen content, nearly 7-fold higher dead cell accumulation, and 2-fold larger lesion area. Hematopoietic SR-BI deletion elicited a maladaptive inflammatory response [higher interleukin (IL)-1ß, IL-6, and TNF-α lower IL-10 and transforming growth factor ß]. Efferocytosis of apoptotic thymocytes was reduced by 64% in SR-BI(-/-) versus WT macrophages, both in vitro and in vivo. In response to apoptotic cells, macrophage SR-BI bound with phosphatidylserine and induced Src phosphorylation and cell membrane recruitment, which led to downstream activation of phosphoinositide 3-kinase (PI3K) and Ras-related C3 botulinum toxin substrate 1 (Rac1) for engulfment and clearance of apoptotic cells, as inhibition of Src decreased PI3K, Rac1-GTP, and efferocytosis in WT cells. Pharmacological inhibition of Rac1 reduced macrophage efferocytosis in a SR-BI-dependent fashion, and activation of Rac1 corrected the defective efferocytosis in SR-BI(-/-) macrophages. Thus, deficiency of macrophage SR-BI promotes defective efferocytosis signaling via the Src/PI3K/Rac1 pathway, resulting in increased plaque size, necrosis, and inflammation.


Assuntos
Aterosclerose/metabolismo , Aterosclerose/patologia , Antígenos CD36/metabolismo , Macrófagos/metabolismo , Fagocitose , Transdução de Sinais , Animais , Apoptose , Aterosclerose/imunologia , Antígenos CD36/deficiência , Antígenos CD36/genética , Sobrevivência Celular , Colágeno/metabolismo , Deleção de Genes , Hematopoese , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Necrose , Fagossomos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilserinas/metabolismo , Transporte Proteico , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 34(12): 2527-36, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25278290

RESUMO

OBJECTIVE: Atherosclerosis is the primary driver of cardiovascular disease, the leading cause of death worldwide. Identification of naturally occurring atheroprotective genes has become a major goal for the development of interventions that will limit atheroma progression and associated adverse events. To this end, we have identified small proline-rich repeat protein (SPRR3) as selectively upregulated in vascular smooth muscle cells (VSMCs) of atheroma-bearing arterial tissue versus healthy arterial tissue. In this study, we sought to determine the role of SPRR3 in atheroma pathophysiology. APPROACH AND RESULTS: We found that atheroprone apolipoprotein E-null mice lacking SPRR3 developed significantly greater atheroma burden. To determine the cellular driver(s) of this increase, we evaluated SPRR3-dependent changes in bone marrow-derived cells, endothelial cells, and VSMCs. Bone marrow transplant of SPRR3-expressing cells into SPRR3(-/-)apolipoprotein E-deficient recipients failed to rescue atheroma burden. Similarly, endothelial cells did not exhibit a response to SPRR3 loss. However, atheromas from SPRR3-deficient mice exhibited increased TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling)-positive VSMCs compared with control. Cell death in SPRR3-deficient VSMCs was significantly increased in vitro. Conversely, SPRR3-overexpressing VSMCs exhibited reduced apoptosis compared with control. We also observed a PI3K (phosphatidylinositol 3-kinase)/Akt-dependent positive association between SPRR3 expression and levels of active Akt in VSMCs. The survival advantage seen in SPRR3-overexpressing VSMCs was abrogated after the addition of a PI3K/Akt pathway inhibitor. CONCLUSIONS: These results indicate that SPRR3 protects the lesion from VSMC loss by promoting survival signaling in plaque VSMCs, thereby significantly decreasing atherosclerosis progression. As the first identified atheroma-specific VSMC prosurvival factor, SPRR3 represents a potential target for lesion-specific modulation of VSMC survival.


Assuntos
Proteínas Ricas em Prolina do Estrato Córneo/metabolismo , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adaptação Fisiológica , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Apoptose , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Proliferação de Células , Sobrevivência Celular , Proteínas Ricas em Prolina do Estrato Córneo/deficiência , Proteínas Ricas em Prolina do Estrato Córneo/genética , Progressão da Doença , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Fosforilação , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Transdução de Sinais
10.
J Lipid Res ; 55(11): 2296-308, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25240046

RESUMO

Macrophages play crucial roles in the formation of atherosclerotic lesions. Akt, a serine/threonine protein kinase B, is vital for cell proliferation, migration, and survival. Macrophages express three Akt isoforms, Akt1, Akt2, and Akt3, but the roles of Akt1 and Akt2 in atherosclerosis in vivo remain unclear. To dissect the impact of macrophage Akt1 and Akt2 on early atherosclerosis, we generated mice with hematopoietic deficiency of Akt1 or Akt2. After 8 weeks on Western diet, Ldlr(-/-) mice reconstituted with Akt1(-/-) fetal liver cells (Akt1(-/-)→Ldlr(-/-)) had similar atherosclerotic lesion areas compared with control mice transplanted with WT cells (WT→Ldlr(-/-)). In contrast, Akt2(-/-)→Ldlr(-/-) mice had dramatically reduced atherosclerotic lesions compared with WT→Ldlr(-/-) mice of both genders. Similarly, in the setting of advanced atherosclerotic lesions, Akt2(-/-)→Ldlr(-/-) mice had smaller aortic lesions compared with WT→Ldlr(-/-) and Akt1(-/-)→Ldlr(-/-) mice. Importantly, Akt2(-/-)→Ldlr(-/-) mice had reduced numbers of proinflammatory blood monocytes expressing Ly-6C(hi) and chemokine C-C motif receptor 2. Peritoneal macrophages isolated from Akt2(-/-) mice were skewed toward an M2 phenotype and showed decreased expression of proinflammatory genes and reduced cell migration. Our data demonstrate that loss of Akt2 suppresses the ability of macrophages to undergo M1 polarization reducing both early and advanced atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de LDL/deficiência , Animais , Antígenos Ly/genética , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/imunologia , Movimento Celular , Feminino , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Hematopoese , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Fenótipo , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores de LDL/genética
11.
Arterioscler Thromb Vasc Biol ; 31(6): 1283-90, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21474828

RESUMO

OBJECTIVE: The adipocyte/macrophage fatty acid-binding proteins aP2 (FABP4) and Mal1 (FABP5) are intracellular lipid chaperones that modulate systemic glucose metabolism, insulin sensitivity, and atherosclerosis. Combined deficiency of aP2 and Mal1 has been shown to reduce the development of atherosclerosis, but the independent role of macrophage Mal1 expression in atherogenesis remains unclear. METHODS AND RESULTS: We transplanted wild-type (WT), Mal1(-/-), or aP2(-/-) bone marrow into low-density lipoprotein receptor-null (LDLR(-/-)) mice and fed them a Western diet for 8 weeks. Mal1(-/-)→LDLR(-/-) mice had significantly reduced (36%) atherosclerosis in the proximal aorta compared with control WT→LDLR(-/-) mice. Interestingly, peritoneal macrophages isolated from Mal1-deficient mice displayed increased peroxisome proliferator-activated receptor-γ (PPARγ) activity and upregulation of a PPARγ-related cholesterol trafficking gene, CD36. Mal1(-/-) macrophages showed suppression of inflammatory genes, such as COX2 and interleukin 6. Mal1(-/-)→LDLR(-/-) mice had significantly decreased macrophage numbers in the aortic atherosclerotic lesions compared with WT→LDLR(-/-) mice, suggesting that monocyte recruitment may be impaired. Indeed, blood monocytes isolated from Mal1(-/-)→LDLR(-/-) mice on a high-fat diet had decreased CC chemokine receptor 2 gene and protein expression levels compared with WT monocytes. CONCLUSION: Taken together, our results demonstrate that Mal1 plays a proatherogenic role by suppressing PPARγ activity, which increases expression of CC chemokine receptor 2 by monocytes, promoting their recruitment to atherosclerotic lesions.


Assuntos
Aterosclerose/prevenção & controle , Proteínas de Ligação a Ácido Graxo/fisiologia , Regulação da Expressão Gênica , Macrófagos/fisiologia , Proteínas de Neoplasias/fisiologia , PPAR gama/fisiologia , Receptores de LDL/fisiologia , Animais , Antígenos CD36/fisiologia , Feminino , Lipídeos/sangue , Camundongos , Receptores CCR2/genética
12.
Am J Physiol Renal Physiol ; 300(5): F1203-13, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21367915

RESUMO

The mechanisms for increased risk of chronic kidney disease (CKD) in obesity remain unclear. The renin-angiotensin system is implicated in the pathogenesis of both adiposity and CKD. We investigated whether the angiotensin type 1 (AT(1)) receptor, composed of dominant AT(1a) and less expressed AT(1b) in wild-type (WT) mice, modulates development and progression of kidney injury in a high-fat diet (HFD)-induced obesity model. WT mice had increased body weight, body fat, and insulin levels and decreased adiponectin levels after 24 wk of a high-fat diet. Identically fed AT(1a) knockout (AT1aKO) mice gained weight similarly to WT mice, but had lower body fat and higher plasma cholesterol. Both obese AT1aKO and obese WT mice had increased visceral fat and kidney macrophage infiltration, with more proinflammatory M1 macrophage markers as well as increased mesangial expansion and tubular vacuolization, compared with lean mice. These abnormalities were heightened in the obese AT1aKO mice, with downregulated M2 macrophage markers and increased macrophage AT(1b) receptor. Treatment with an AT(1) receptor blocker, which affects both AT(1a) and AT(1b), abolished renal macrophage infiltration with inhibition of renal M1 and upregulation of M2 macrophage markers in obese WT mice. Our data suggest obesity accelerates kidney injury, linked to augmented inflammation in adipose and kidney tissues and a proinflammatory shift in macrophage and M1/M2 balance.


Assuntos
Gordura Intra-Abdominal/metabolismo , Nefropatias/prevenção & controle , Rim/metabolismo , Macrófagos/metabolismo , Obesidade/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Adiponectina/sangue , Adiposidade , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Biomarcadores/sangue , Peso Corporal , Colesterol/sangue , Gorduras na Dieta , Modelos Animais de Doenças , Hemodinâmica , Mediadores da Inflamação/sangue , Insulina/sangue , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/imunologia , Gordura Intra-Abdominal/patologia , Rim/efeitos dos fármacos , Rim/imunologia , Rim/patologia , Nefropatias/etiologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/complicações , Obesidade/genética , Obesidade/imunologia , Obesidade/patologia , Receptor Tipo 1 de Angiotensina/deficiência , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptor Tipo 1 de Angiotensina/genética
13.
J Clin Invest ; 121(4): 1624-35, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21383501

RESUMO

Although the response of endothelial cells to the disturbed blood flow in the vicinity of atherosclerotic lesions is known to be distinct from that elicited by nonatherogenic laminar flow, the mechanisms involved are poorly understood. Our initial studies confirmed that expression of the endothelial receptor tyrosine kinase Tie1 was evident at regions of atherogenic flow in mature animals. We therefore hypothesized that Tie1 plays a role in the endothelial response to atherogenic shear stress. Consistent with this, we found that Tie1+/- mice bred to the apoE-deficient background displayed a 35% reduction in atherosclerosis relative to Tie1+/+;Apoe-/- mice. Since deletion of Tie1 results in embryonic lethality secondary to vascular dysfunction, we used conditional and inducible mutagenesis to study the effect of endothelial-specific Tie1 attenuation on atherogenesis in Apoe-/- mice and found a dose-dependent decrease in atherosclerotic lesions. Analysis of primary aortic endothelial cells indicated that atheroprotective laminar flow decreased Tie1 expression in vitro. Attenuation of Tie1 was associated with an increase in eNOS expression and Tie2 phosphorylation. In addition, Tie1 attenuation increased IkBα expression while decreasing ICAM levels. In summary, we have found that shear stress conditions that modulate atherogenic events also regulate Tie1 expression. Therefore, Tie1 may play a novel proinflammatory role in atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Receptor de TIE-1/deficiência , Receptor de TIE-1/genética , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/fisiopatologia , Sequência de Bases , Moléculas de Adesão Celular/genética , Primers do DNA/genética , Modelos Animais de Doenças , Células Endoteliais/fisiologia , Feminino , Expressão Gênica , Hemorreologia , Óperon Lac , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Óxido Nítrico Sintase Tipo III/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de TIE-1/fisiologia , Transdução de Sinais , Estresse Mecânico , Quinases Associadas a rho/genética
14.
Free Radic Biol Med ; 50(1): 27-36, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20974251

RESUMO

To test whether severe ascorbic acid deficiency in macrophages affects progression of early atherosclerosis, we used fetal liver cell transplantation to generate atherosclerosis-prone apolipoprotein E-deficient (apoE(-/-)) mice that selectively lacked the ascorbate transporter (SVCT2) in hematopoietic cells, including macrophages. After 13 weeks of chow diet, apoE(-/-) mice lacking the SVCT2 in macrophages had surprisingly less aortic atherosclerosis, decreased lesion macrophage numbers, and increased macrophage apoptosis compared to control-transplanted mice. Serum lipid levels were similar in both groups. Peritoneal macrophages lacking the SVCT2 had undetectable ascorbate; increased susceptibility to H(2)O(2)-induced mitochondrial dysfunction and apoptosis; decreased expression of genes for COX-2, IL1ß, and IL6; and decreased lipopolysaccharide-stimulated NF-κB and antiapoptotic gene expression. These changes were associated with decreased expression of both the receptor for advanced glycation end products and HIF-1α, either or both of which could have been the proximal cause of decreased macrophage activation and apoptosis in ascorbate-deficient macrophages.


Assuntos
Deficiência de Ácido Ascórbico/genética , Aterosclerose/genética , Aterosclerose/prevenção & controle , Macrófagos Peritoneais/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores/genética , Animais , Apolipoproteínas E/genética , Ácido Ascórbico/efeitos adversos , Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/metabolismo , Deficiência de Ácido Ascórbico/patologia , Aterosclerose/etiologia , Aterosclerose/patologia , Células Cultivadas , Progressão da Doença , Feminino , Predisposição Genética para Doença/prevenção & controle , Macrófagos Peritoneais/patologia , Macrófagos Peritoneais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/fisiologia , Transportadores de Sódio Acoplados à Vitamina C , Simportadores/metabolismo , Simportadores/fisiologia , Fatores de Tempo
15.
Arterioscler Thromb Vasc Biol ; 30(9): 1751-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20558818

RESUMO

OBJECTIVE: To assess the role of combined deficiencies of vitamins C and E on the earliest stages of atherosclerosis (an inflammatory condition associated with oxidative stress), 4 combinations of vitamin supplementation (low C/low E, low C/high E, high C/low E, and high C/high E) were studied in atherosclerosis-prone apolipoprotein E-deficient mice also unable to synthesize their own vitamin C (gulonolactone oxidase(-/-)); and to evaluate the effect of a more severe depletion of vitamin C alone in a second experiment using gulonolactone oxidase(-/-) mice carrying the hemizygous deletion of SVCT2 (the vitamin C transporter). METHODS AND RESULTS: After 8 weeks of a high-fat diet (16% lard and 0.2% cholesterol), atherosclerosis developed in the aortic sinus areas of mice in all diet groups. Each vitamin-deficient diet significantly decreased liver and brain contents of the corresponding vitamin. Combined deficiency of both vitamins increased lipid peroxidation, doubled plaque size, and increased plaque macrophage content by 2- to 3-fold in male mice, although only plaque macrophage content was increased in female mice. A more severe deficiency of vitamin C in gulonolactone oxidase(-/-) mice with defective cellular uptake of vitamin C increased both oxidative stress and atherosclerosis in apolipoprotein E(-/-) mice compared with littermates receiving a diet replete in vitamin C, again most clearly in males. CONCLUSIONS: Combined deficiencies of vitamins E and C are required to worsen early atherosclerosis in an apolipoprotein E-deficient mouse model. However, a more severe cellular deficiency of vitamin C alone promotes atherosclerosis when vitamin E is replete.


Assuntos
Doenças da Aorta/etiologia , Apolipoproteínas E/deficiência , Deficiência de Ácido Ascórbico/complicações , Aterosclerose/etiologia , Deficiência de Vitamina E/complicações , Animais , Doenças da Aorta/tratamento farmacológico , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Ácido Ascórbico/metabolismo , Ácido Ascórbico/farmacologia , Deficiência de Ácido Ascórbico/tratamento farmacológico , Deficiência de Ácido Ascórbico/metabolismo , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Encéfalo/metabolismo , Suplementos Nutricionais , Modelos Animais de Doenças , Progressão da Doença , Feminino , L-Gulonolactona Oxidase/deficiência , L-Gulonolactona Oxidase/genética , Peroxidação de Lipídeos , Fígado/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/deficiência , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Estresse Oxidativo , Índice de Gravidade de Doença , Fatores Sexuais , Transportadores de Sódio Acoplados à Vitamina C , Simportadores/deficiência , Simportadores/genética , Fatores de Tempo , Vitamina E/metabolismo , Vitamina E/farmacologia , Deficiência de Vitamina E/tratamento farmacológico , Deficiência de Vitamina E/metabolismo , Vitaminas/metabolismo , Vitaminas/farmacologia
16.
Nat Med ; 15(12): 1383-91, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19966778

RESUMO

Macrophages show endoplasmic reticulum (ER) stress when exposed to lipotoxic signals associated with atherosclerosis, although the pathophysiological importance and the underlying mechanisms of this phenomenon remain unknown. Here we show that mitigation of ER stress with a chemical chaperone results in marked protection against lipotoxic death in macrophages and prevents macrophage fatty acid-binding protein-4 (aP2) expression. Using genetic and chemical models, we show that aP2 is the predominant regulator of lipid-induced macrophage ER stress. The absence of lipid chaperones incites an increase in the production of phospholipids rich in monounsaturated fatty acids and bioactive lipids that render macrophages resistant to lipid-induced ER stress. Furthermore, the impact of aP2 on macrophage lipid metabolism and the ER stress response is mediated by upregulation of key lipogenic enzymes by the liver X receptor. Our results demonstrate the central role for lipid chaperones in regulating ER homeostasis in macrophages in atherosclerosis and show that ER responses can be modified, genetically or chemically, to protect the organism against the deleterious effects of hyperlipidemia.


Assuntos
Aterosclerose/prevenção & controle , Retículo Endoplasmático/metabolismo , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Humanos , Estresse Oxidativo
17.
Cell Metab ; 8(6): 492-501, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19041765

RESUMO

Prostaglandin (PG) E(2), a major product of activated macrophages, has been implicated in atherosclerosis and plaque rupture. The PGE(2) receptors, EP2 and EP4, are expressed in atherosclerotic lesions and are known to inhibit apoptosis in cancer cells. To examine the roles of macrophage EP4 and EP2 in apoptosis and early atherosclerosis, fetal liver cell transplantation was used to generate LDLR(-/-) mice chimeric for EP2(-/-) or EP4(-/-) hematopoietic cells. After 8 weeks on a Western diet, EP4(-/-) --> LDLR(-/-) mice, but not EP2(-/-) --> LDLR(-/-) mice, had significantly reduced aortic atherosclerosis with increased apoptotic cells in the lesions. EP4(-/-) peritoneal macrophages had increased sensitivity to proapoptotic stimuli, including palmitic acid and free cholesterol loading, which was accompanied by suppression of activity of p-Akt, p-Bad, and NF-kappaB-regulated genes. Thus, EP4 deficiency inhibits the PI3K/Akt and NF-kappaB pathways compromising macrophage survival and suppressing early atherosclerosis, identifying macrophage EP4-signaling pathways as molecular targets for modulating the development of atherosclerosis.


Assuntos
Apoptose , Aterosclerose/etiologia , Macrófagos/metabolismo , Receptores de Prostaglandina E/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Células Cultivadas , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP4 , Transdução de Sinais/genética
18.
Atherosclerosis ; 197(2): 534-40, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17897651

RESUMO

Atherosclerotic lesions are characterized by a profound alteration in the architecture of the arterial intima, with a marked increase of fibronectin (FN) and the appearance of the alternatively spliced FN variant containing the extra domain A (EDA). To analyze the role of FN isoforms in atherosclerotic lesion formation we utilized mouse strains devoid of EDA exon regulated splicing, which constitutively include (EDA(+/+)) or exclude (EDA(-/-)) the exon. Both mutant mice had a 40% reduction in atherosclerotic lesions after the atherogenic-diet treatment (mean+/-S.E., microm(2); 22969+/-2185; 13660+/-1533; 14260+/-2501 for EDA(wt/wt), EDA(+/+) and EDA(-/-), respectively; p< or =0.01 ANOVA test) associated to a lower capacity of macrophages to uptake modified LDL and undergo foam-cell formation. Lesions in control mice were more numerous and bigger, with augmented and deeper macrophage infiltration, and increased FN expression in the sub-endothelial area. Previous experiments have shown that apoE(-/-)EDA(-/-) mice have a decreased number and size of atherosclerotic lesions and, on this basis, it has been proposed that the EDA domain has a pro-atherogenic role. Our data with the EDA(+/+) mice rules out this hypothesis and suggest that regulated splicing of the EDA exon of the FN gene is involved in progression of atherosclerosis, highlighting the importance of alternative splicing in regulating cellular processes.


Assuntos
Processamento Alternativo/fisiologia , Aterosclerose/fisiopatologia , Éxons/genética , Fibronectinas/genética , Fibronectinas/fisiologia , Processamento Alternativo/genética , Animais , Dieta Aterogênica , Modelos Animais de Doenças , Feminino , Células Espumosas , Camundongos , Isoformas de Proteínas/genética
19.
Circulation ; 116(12): 1404-12, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17724261

RESUMO

BACKGROUND: The peroxisome proliferator-activated receptor-alpha (PPARalpha) plays important roles in lipid metabolism, inflammation, and atherosclerosis. PPARalpha ligands have been shown to reduce cardiovascular events in high-risk subjects. PPARalpha expression by arterial cells, including macrophages, may exert local antiatherogenic effects independent of plasma lipid changes. METHODS AND RESULTS: To examine the contribution of PPARalpha expression by bone marrow-derived cells in atherosclerosis, male and female low-density lipoprotein receptor-deficient (LDLR(-/-)) mice were reconstituted with bone marrow from PPARalpha(-/-) or PPARalpha(+/+) mice and challenged with a high-fat diet. Although serum lipids and lipoprotein profiles did not differ between the groups, the size of atherosclerotic lesions in the distal aorta of male and female PPARalpha(-/-) --> LDLR(-/-) mice was significantly increased (44% and 46%, respectively) compared with controls. Male PPARalpha(-/-) --> LDLR(-/-) mice also had larger (44%) atherosclerotic lesions in the proximal aorta than male PPARalpha(+/+) --> LDLR(-/-) mice. Peritoneal macrophages from PPARalpha(-/-) mice had increased uptake of oxidized LDL and decreased cholesterol efflux. PPARalpha(-/-) macrophages had lower levels of scavenger receptor B type I and ABCA1 protein expression and an accelerated response of nuclear factor-kappaB-regulated inflammatory genes. A laser capture microdissection analysis verified suppressed scavenger receptor B type I and increased nuclear factor-kappaB gene expression levels in vivo in atherosclerotic lesions of PPARalpha(-/-) --> LDLR(-/-) mice compared with the lesions of control PPARalpha(+/+) --> LDLR(-/-) mice. CONCLUSIONS: These data demonstrate that PPARalpha expression by macrophages has antiatherogenic effects via modulation of cell cholesterol trafficking and inflammatory activity.


Assuntos
Aterosclerose/prevenção & controle , Macrófagos/metabolismo , PPAR alfa/fisiologia , Receptores de LDL/deficiência , Animais , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Doenças da Aorta/prevenção & controle , Aterosclerose/metabolismo , Aterosclerose/patologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Transplante de Medula Óssea , Butiratos/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR alfa/agonistas , PPAR alfa/deficiência , PPAR alfa/genética , Compostos de Fenilureia/farmacologia , Pirimidinas/farmacologia , Quimera por Radiação , Receptores de LDL/genética
20.
J Am Soc Nephrol ; 18(8): 2311-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17634441

RESUMO

Although increased extracellular matrix (ECM) is pathogenic in a variety of chronic tissue injuries, reduced and/or disrupted ECM may be detrimental in atherosclerosis and rather destabilize existing atherosclerotic lesions. This study therefore assessed the effects of angiotensin II (AngII) antagonism on ECM components of advanced atherosclerosis. Twenty-four-week-old apolipoprotein E-deficient mice were treated with the AngII antagonist losartan for 12 wk. Controls received water or hydralazine. AngII antagonism significantly reduced progression of established atherosclerosis, whereas hydralazine showed no benefit despite similar decrease in BP. Although there was no difference in the macrophage component, AngII antagonism increased the relative collagen portion of the lesions; lessened elastin fragmentation, increased the total elastin content of the aorta; and reduced the mRNA and activity/protein of the elastolytic proteases, cathepsin S, and metalloproteinase-9. Extracellular elastin degradation by cultured smooth muscle cells (SMC) was reduced by losartan, as was SMC invasion through an elastin gel barrier. Thus, AngII antagonism lessens progression of atherosclerosis, increases collagen, and preserves elastin components of ECM within the vascular lesions, which, at least in part, is modulated by effects on SMC. These effects not only decrease further expansion of advanced lesions but also stabilize the established atherosclerotic plaques and may underlie the decreased incidence of acute cardiovascular events that are observed in patients in whom AngII antagonism is begun after atherosclerosis is already established.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Losartan/farmacologia , Angiotensina II/sangue , Angiotensina II/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Doenças da Aorta/tratamento farmacológico , Doenças da Aorta/patologia , Apolipoproteínas E/genética , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Progressão da Doença , Elasticidade , Feminino , Hidralazina/farmacologia , Lipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Vasoconstritores/sangue , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...