Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 205(4): 1070-1083, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32661179

RESUMO

IL-4 activates macrophages to adopt distinct phenotypes associated with clearance of helminth infections and tissue repair, but the phenotype depends on the cellular lineage of these macrophages. The molecular basis of chromatin remodeling in response to IL-4 stimulation in tissue-resident and monocyte-derived macrophages is not understood. In this study, we find that IL-4 activation of different lineages of peritoneal macrophages in mice is accompanied by lineage-specific chromatin remodeling in regions enriched with binding motifs of the pioneer transcription factor PU.1. PU.1 motif is similarly associated with both tissue-resident and monocyte-derived IL-4-induced accessible regions but has different lineage-specific DNA shape features and predicted cofactors. Mutation studies based on natural genetic variation between C57BL/6 and BALB/c mouse strains indicate that accessibility of these IL-4-induced regions can be regulated through differences in DNA shape without direct disruption of PU.1 motifs. We propose a model whereby DNA shape features of stimulation-dependent genomic elements contribute to differences in the accessible chromatin landscape of alternatively activated macrophages on different genetic backgrounds that may contribute to phenotypic variations in immune responses.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Cromatina/genética , DNA/genética , Macrófagos Peritoneais/fisiologia , Proteínas Proto-Oncogênicas/genética , Transativadores/genética , Animais , Sítios de Ligação/genética , Imunidade/genética , Interleucina-4/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Mutação/genética , Ligação Proteica/genética
2.
Cell Host Microbe ; 25(3): 463-470.e9, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30799265

RESUMO

The pathogenesis of Staphylococcus aureus is thought to depend on the production of pore-forming leukocidins that kill leukocytes and lyse erythrocytes. Two leukocidins, Leukocidin ED (LukED) and γ-Hemolysin AB (HlgAB), are necessary and sufficient to kill mice upon infection and toxin challenge. We demonstrate that LukED and HlgAB cause vascular congestion and derangements in vascular fluid distribution that rapidly cause death in mice. The Duffy antigen receptor for chemokines (DARC) on endothelial cells, rather than leukocytes or erythrocytes, is the critical target for lethality. Consistent with this, LukED and HlgAB injure primary human endothelial cells in a DARC-dependent manner, and mice with DARC-deficient endothelial cells are resistant to toxin-mediated lethality. During bloodstream infection in mice, DARC targeting by S. aureus causes increased tissue damage, organ dysfunction, and host death. The potential for S. aureus leukocidins to manipulate vascular integrity highlights the importance of these virulence factors.


Assuntos
Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Sistema do Grupo Sanguíneo Duffy/metabolismo , Células Endoteliais/efeitos dos fármacos , Exotoxinas/toxicidade , Proteínas Hemolisinas/toxicidade , Receptores de Superfície Celular/metabolismo , Infecções Estafilocócicas/patologia , Staphylococcus aureus/patogenicidade , Animais , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Exotoxinas/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Staphylococcus aureus/metabolismo , Análise de Sobrevida
3.
Cancer Immunol Res ; 5(12): 1109-1121, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29097420

RESUMO

Efficacious antitumor immune responses must overcome multiple suppressive mechanisms in the tumor microenvironment to control cancer progression. In this study, we demonstrate that dual targeting of suppressive myeloid populations by inhibiting CSF-1/CSF-1R signaling and activation of antigen-presenting cells with agonist anti-CD40 treatment confers superior antitumor efficacy and increased survival compared with monotherapy treatment in preclinical tumor models. Concurrent CSF-1R blockade and CD40 agonism lead to profound changes in the composition of immune infiltrates, causing an overall decrease in immunosuppressive cells and a shift toward a more inflammatory milieu. Anti-CD40/anti-CSF-1R-treated tumors contain decreased tumor-associated macrophages and Foxp3+ regulatory T cells. This combination approach increases maturation and differentiation of proinflammatory macrophages and dendritic cells and also drives potent priming of effector T cells in draining lymph nodes. As a result, tumor-infiltrating effector T cells exhibit improved responses to tumor antigen rechallenge. These studies show that combining therapeutic approaches may simultaneously remove inhibitory immune populations and sustain endogenous antitumor immune responses to successfully impair cancer progression. Cancer Immunol Res; 5(12); 1109-21. ©2017 AACR.


Assuntos
Antineoplásicos/farmacologia , Antígenos CD40/antagonistas & inibidores , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/farmacologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Camundongos , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
4.
J Clin Invest ; 127(8): 2904-2915, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28650342

RESUMO

Atherosclerosis is a chronic inflammatory disease, and developing therapies to promote its regression is an important clinical goal. We previously established that atherosclerosis regression is characterized by an overall decrease in plaque macrophages and enrichment in markers of alternatively activated M2 macrophages. We have now investigated the origin and functional requirement for M2 macrophages in regression in normolipidemic mice that received transplants of atherosclerotic aortic segments. We compared plaque regression in WT normolipidemic recipients and those deficient in chemokine receptors necessary to recruit inflammatory Ly6Chi (Ccr2-/- or Cx3cr1-/-) or patrolling Ly6Clo (Ccr5-/-) monocytes. Atherosclerotic plaques transplanted into WT or Ccr5-/- recipients showed reduced macrophage content and increased M2 markers consistent with plaque regression, whereas plaques transplanted into Ccr2-/- or Cx3cr1-/- recipients lacked this regression signature. The requirement of recipient Ly6Chi monocyte recruitment was confirmed in cell trafficking studies. Fate-mapping and single-cell RNA sequencing studies also showed that M2-like macrophages were derived from newly recruited monocytes. Furthermore, we used recipient mice deficient in STAT6 to demonstrate a requirement for this critical component of M2 polarization in atherosclerosis regression. Collectively, these results suggest that continued recruitment of Ly6Chi inflammatory monocytes and their STAT6-dependent polarization to the M2 state are required for resolution of atherosclerotic inflammation and plaque regression.


Assuntos
Aterosclerose/sangue , Macrófagos/citologia , Monócitos/citologia , Placa Aterosclerótica/sangue , Animais , Aorta/metabolismo , Aorta/fisiologia , Aterosclerose/metabolismo , Receptor 1 de Quimiocina CX3C , Linhagem da Célula , Feminino , Hiperlipidemias/sangue , Inflamação , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Fenótipo , Placa Aterosclerótica/metabolismo , Receptores CCR2/genética , Receptores CCR5/genética , Receptores de Quimiocinas/genética , Fator de Transcrição STAT6/metabolismo , Análise de Sequência de RNA , Resultado do Tratamento
5.
Nat Immunol ; 18(6): 642-653, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28436955

RESUMO

It remains unclear whether activated inflammatory macrophages can adopt features of tissue-resident macrophages, or what mechanisms might mediate such a phenotypic conversion. Here we show that vitamin A is required for the phenotypic conversion of interleukin 4 (IL-4)-activated monocyte-derived F4/80intCD206+PD-L2+MHCII+ macrophages into macrophages with a tissue-resident F4/80hiCD206-PD-L2-MHCII-UCP1+ phenotype in the peritoneal cavity of mice and during the formation of liver granulomas in mice infected with Schistosoma mansoni. The phenotypic conversion of F4/80intCD206+ macrophages into F4/80hiCD206- macrophages was associated with almost complete remodeling of the chromatin landscape, as well as alteration of the transcriptional profiles. Vitamin A-deficient mice infected with S. mansoni had disrupted liver granuloma architecture and increased mortality, which indicates that failure to convert macrophages from the F4/80intCD206+ phenotype to F4/80hiCD206- may lead to dysregulated inflammation during helminth infection.


Assuntos
Granuloma/imunologia , Fígado/imunologia , Macrófagos/imunologia , Esquistossomose mansoni/imunologia , Deficiência de Vitamina A/imunologia , Animais , Antígenos de Diferenciação/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/metabolismo , Interleucina-4/imunologia , Lectinas Tipo C/metabolismo , Fígado/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Cavidade Peritoneal/citologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular/metabolismo , Schistosoma mansoni , Esquistossomose mansoni/patologia , Tretinoína/farmacologia , Proteína Desacopladora 1/metabolismo , Vitaminas/farmacologia
6.
PLoS Pathog ; 10(12): e1004528, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25474413

RESUMO

Cerebral malaria claims the lives of over 600,000 African children every year. To better understand the pathogenesis of this devastating disease, we compared the cellular dynamics in the cortical microvasculature between two infection models, Plasmodium berghei ANKA (PbA) infected CBA/CaJ mice, which develop experimental cerebral malaria (ECM), and P. yoelii 17XL (PyXL) infected mice, which succumb to malarial hyperparasitemia without neurological impairment. Using a combination of intravital imaging and flow cytometry, we show that significantly more CD8(+) T cells, neutrophils, and macrophages are recruited to postcapillary venules during ECM compared to hyperparasitemia. ECM correlated with ICAM-1 upregulation on macrophages, while vascular endothelia upregulated ICAM-1 during ECM and hyperparasitemia. The arrest of large numbers of leukocytes in postcapillary and larger venules caused microrheological alterations that significantly restricted the venous blood flow. Treatment with FTY720, which inhibits vascular leakage, neurological signs, and death from ECM, prevented the recruitment of a subpopulation of CD45(hi) CD8(+) T cells, ICAM-1(+) macrophages, and neutrophils to postcapillary venules. FTY720 had no effect on the ECM-associated expression of the pattern recognition receptor CD14 in postcapillary venules suggesting that endothelial activation is insufficient to cause vascular pathology. Expression of the endothelial tight junction proteins claudin-5, occludin, and ZO-1 in the cerebral cortex and cerebellum of PbA-infected mice with ECM was unaltered compared to FTY720-treated PbA-infected mice or PyXL-infected mice with hyperparasitemia. Thus, blood brain barrier opening does not involve endothelial injury and is likely reversible, consistent with the rapid recovery of many patients with CM. We conclude that the ECM-associated recruitment of large numbers of activated leukocytes, in particular CD8(+) T cells and ICAM(+) macrophages, causes a severe restriction in the venous blood efflux from the brain, which exacerbates the vasogenic edema and increases the intracranial pressure. Thus, death from ECM could potentially occur as a consequence of intracranial hypertension.


Assuntos
Barreira Hematoencefálica/imunologia , Córtex Cerebral/imunologia , Malária Cerebral/imunologia , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , Animais , Barreira Hematoencefálica/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Córtex Cerebral/parasitologia , Córtex Cerebral/patologia , Claudina-5/imunologia , Modelos Animais de Doenças , Cloridrato de Fingolimode , Humanos , Imunossupressores/farmacologia , Molécula 1 de Adesão Intercelular/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Malária Cerebral/tratamento farmacológico , Malária Cerebral/patologia , Camundongos , Neutrófilos/imunologia , Neutrófilos/patologia , Ocludina/imunologia , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Proteína da Zônula de Oclusão-1/imunologia
7.
Circ Res ; 115(9): 759-69, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25201910

RESUMO

RATIONALE: Diabetes mellitus increases cardiovascular disease risk in humans and remains elevated despite cholesterol-lowering therapy with statins. Consistent with this, in mouse models, diabetes mellitus impairs atherosclerosis plaque regression after aggressive cholesterol lowering. MicroRNA 33 (miR33) is a key negative regulator of the reverse cholesterol transport factors, ATP-binding cassette transporter A1 and high-density lipoprotein, which suggested that its inhibition may overcome this impairment. OBJECTIVE: To assess the effects of miR33 inhibition on atherosclerosis regression in diabetic mice. METHODS AND RESULTS: Reversa mice, which are deficient in the low-density lipoprotein receptor and in which hypercholesterolemia is reversed by conditional inactivation of the microsomal triglyceride transfer protein gene, were placed on an atherogenic diet for 16 weeks, then either made diabetic by streptozotocin injection or kept normoglycemic. Lipid-lowering was induced by microsomal triglyceride transfer protein gene inactivation, and mice were treated with anti-miR33 or control oligonucleotides. Although regression was impaired in diabetic mice treated with control oligonucleotides, anti-miR33 treatment decreased plaque macrophage content and inflammatory gene expression in these mice. The decreased macrophage content in anti-miR33 treated diabetic mice was associated with a blunting of hyperglycemia-induced monocytosis and reduced monocyte recruitment to the plaque, which was traced to an inhibition of the proliferation of bone marrow monocyte precursors associated with the upregulation of their Abca1. CONCLUSIONS: miR33 inhibition overcomes deleterious effects of diabetes mellitus in atherosclerosis regression in mice, which suggests a therapeutic strategy in diabetic patients, who remain at elevated cardiovascular disease risk, despite plasma cholesterol lowering.


Assuntos
Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Diabetes Mellitus Experimental/terapia , Angiopatias Diabéticas/prevenção & controle , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos Antissenso/administração & dosagem , Placa Aterosclerótica , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Doenças da Aorta/sangue , Doenças da Aorta/etiologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/sangue , Aterosclerose/etiologia , Aterosclerose/genética , Aterosclerose/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Angiopatias Diabéticas/sangue , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/patologia , Dieta Ocidental , Progressão da Doença , Inativação Gênica , Mediadores da Inflamação/metabolismo , Lipídeos/sangue , Lipoproteínas/genética , Lipoproteínas/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Células-Tronco/metabolismo , Fatores de Tempo
8.
PLoS Pathog ; 10(6): e1004080, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24967715

RESUMO

Alternatively activated macrophages (AAM) that accumulate during chronic T helper 2 inflammatory conditions may arise through proliferation of resident macrophages or recruitment of monocyte-derived cells. Liver granulomas that form around eggs of the helminth parasite Schistosoma mansoni require AAM to limit tissue damage. Here, we characterized monocyte and macrophage dynamics in the livers of infected CX3CR1(GFP/+) mice. CX3CR1-GFP⁺ monocytes and macrophages accumulated around eggs and in granulomas during infection and upregulated PD-L2 expression, indicating differentiation into AAM. Intravital imaging of CX3CR1-GFP⁺ Ly6C(low) monocytes revealed alterations in patrolling behavior including arrest around eggs that were not encased in granulomas. Differential labeling of CX3CR1-GFP⁺ cells in the blood and the tissue showed CD4⁺ T cell dependent accumulation of PD-L2⁺ CX3CR1-GFP⁺ AAM in the tissues as granulomas form. By adoptive transfer of Ly6C(high) and Ly6C(low) monocytes into infected mice, we found that AAM originate primarily from transferred Ly6C(high) monocytes, but that these cells may transition through a Ly6C(low) state and adopt patrolling behavior in the vasculature. Thus, during chronic helminth infection AAM can arise from recruited Ly6C(high) monocytes via help from CD4⁺ T cells.


Assuntos
Antígenos Ly/sangue , Linfócitos T CD4-Positivos/imunologia , Granuloma/imunologia , Fígado/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Esquistossomose mansoni/imunologia , Animais , Antígenos Ly/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/parasitologia , Comunicação Celular , Transdiferenciação Celular , Cruzamentos Genéticos , Feminino , Granuloma/parasitologia , Granuloma/patologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Vigilância Imunológica , Fígado/metabolismo , Fígado/parasitologia , Fígado/patologia , Ativação de Macrófagos , Macrófagos/metabolismo , Macrófagos/parasitologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/parasitologia , Óvulo/crescimento & desenvolvimento , Óvulo/imunologia , Proteína 2 Ligante de Morte Celular Programada 1/metabolismo , Proteínas Recombinantes/metabolismo , Schistosoma mansoni/crescimento & desenvolvimento , Schistosoma mansoni/imunologia , Esquistossomose mansoni/metabolismo , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/fisiopatologia , Regulação para Cima
9.
Blood ; 123(20): e110-22, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24695852

RESUMO

Macrophages adopt an alternatively activated phenotype (AAMs) when activated by the interleukin-4receptor(R)α. AAMs can be derived either from proliferation of tissue resident macrophages or recruited inflammatory monocytes, but it is not known whether these different sources generate AAMs that are phenotypically and functionally distinct. By transcriptional profiling analysis, we show here that, although both monocyte and tissue-derived AAMs expressed high levels of Arg1, Chi3l3, and Retnla, only monocyte-derived AAMs up-regulated Raldh2 and PD-L2. Monocyte-derived AAMs were also CX3CR1-green fluorescent protein (GFP)(high) and expressed CD206, whereas tissue-derived AAMs were CX3CR1-GFP and CD206 negative. Monocyte-derived AAMs had high levels of aldehyde dehydrogenase activity and promoted the differentiation of FoxP3(+) cells from naïve CD4(+) cells via production of retinoic acid. In contrast, tissue-derived AAMs expressed high levels of uncoupling protein 1. Hence monocyte-derived AAM have properties associated with immune regulation, and the different physiological properties associated with AAM function may depend on the distinct lineage of these cells.


Assuntos
Perfilação da Expressão Gênica , Ativação de Macrófagos , Macrófagos/imunologia , Monócitos/imunologia , Animais , Antígenos CD4/análise , Proliferação de Células , Células Cultivadas , Fatores de Transcrição Forkhead/análise , Expressão Gênica , Canais Iônicos/análise , Canais Iônicos/genética , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/análise , Proteínas Mitocondriais/genética , Monócitos/citologia , Monócitos/metabolismo , Proteína Desacopladora 1
11.
Infect Immun ; 81(5): 1635-43, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23460516

RESUMO

Natural infection by Toxoplasma gondii occurs via oral ingestion of tissue cysts that rupture in the small intestine, releasing zoites that infect locally before disseminating throughout the host. The studies presented here used fluorescent parasites combined with flow cytometry and multiphoton microscopy techniques to understand the events associated with parasite replication in the mucosa. At 3 days postinfection with tissue cysts, parasites were localized in small foci and flow cytometry revealed parasites present in macrophages, neutrophils, and monocytes in the lamina propria. By day 6 postinfection, there were large foci of replicating parasites; however, foci unexpectedly varied in the number of villi involved and were associated with the presence of viable tachyzoites within the intestinal lumen. Consistent with the flow cytometry data, neutrophils and monocytes in the lamina propria were preferentially associated with parasite plaques. In contrast, dendritic cells comprised a small fraction of the infected immune cell population and were localized at the periphery of parasite plaques. Together, these findings reveal the formation of localized sites of parasite replication and inflammation early during infection and suggest that sustained replication of T. gondii in the gut may be a function of pathogen luminal spread.


Assuntos
Intestino Delgado/parasitologia , Toxoplasma/crescimento & desenvolvimento , Toxoplasmose Animal/parasitologia , Animais , Modelos Animais de Doenças , Feminino , Mucosa Intestinal/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Toxoplasma/isolamento & purificação
12.
PLoS Pathog ; 8(8): e1002883, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22927819

RESUMO

Although the vitamin A metabolite retinoic acid (RA) plays a critical role in immune function, RA synthesis during infection is poorly understood. Here, we show that retinal dehydrogenases (Raldh), required for the synthesis of RA, are induced during a retinoid-dependent type-2 immune response elicited by Schistosoma mansoni infection, but not during a retinoid-independent anti-viral immune response. Vitamin A deficient mice have a selective defect in T(H)2 responses to S. mansoni, but retained normal LCMV specific T(H)1 responses. A combination of in situ imaging, intra-vital imaging, and sort purification revealed that alternatively activated macrophages (AAMφ) express high levels of Raldh2 during S. mansoni infection. IL-4 induces Raldh2 expression in bone marrow-derived macrophages in vitro and peritoneal macrophages in vivo. Finally, in vivo derived AAMφ have an enhanced capacity to induce Foxp3 expression in CD4+ cells through an RA dependent mechanism, especially in combination with TGF-ß. The regulation of Raldh enzymes during infection is pathogen specific and reflects differential requirements for RA during effector responses. Specifically, AAMφ are an inducible source of RA synthesis during helminth infections and T(H)2 responses that may be important in regulating immune responses.


Assuntos
Regulação Enzimológica da Expressão Gênica/imunologia , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Retinal Desidrogenase/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Regulação para Cima/imunologia , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação Enzimológica da Expressão Gênica/genética , Ativação de Macrófagos/genética , Camundongos , Camundongos Knockout , Retinal Desidrogenase/biossíntese , Retinal Desidrogenase/genética , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/enzimologia , Esquistossomose mansoni/genética , Células Th1/imunologia , Células Th2/imunologia , Regulação para Cima/genética
13.
PLoS One ; 7(4): e35086, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496894

RESUMO

Monkeypox virus (MPXV) causes a smallpox-like disease in humans. Clinical and epidemiological studies provide evidence of pathogenicity differences between two geographically distinct monkeypox virus clades: the West African and Congo Basin. Genomic analysis of strains from both clades identified a ∼10 kbp deletion in the less virulent West African isolates sequenced to date. One absent open reading frame encodes the monkeypox virus homologue of the complement control protein (CCP). This modulatory protein prevents the initiation of both the classical and alternative pathways of complement activation. In monkeypox virus, CCP, also known as MOPICE, is a ∼24 kDa secretory protein with sequence homology to this superfamily of proteins. Here we investigate CCP expression and its role in monkeypox virulence and pathogenesis. CCP was incorporated into the West African strain and removed from the Congo Basin strain by homologous recombination. CCP expression phenotypes were confirmed for both wild type and recombinant monkeypox viruses and CCP activity was confirmed using a C4b binding assay. To characterize the disease, prairie dogs were intranasally infected and disease progression was monitored for 30 days. Removal of CCP from the Congo Basin strain reduced monkeypox disease morbidity and mortality, but did not significantly decrease viral load. The inclusion of CCP in the West African strain produced changes in disease manifestation, but had no apparent effect on disease-associated mortality. This study identifies CCP as an important immuno-modulatory protein in monkeypox pathogenesis but not solely responsible for the increased virulence seen within the Congo Basin clade of monkeypox virus.


Assuntos
Ativação do Complemento , Monkeypox virus/imunologia , Monkeypox virus/patogenicidade , Mpox/imunologia , Mpox/virologia , Proteínas Virais/imunologia , Animais , Sequência de Bases , Proteína de Ligação ao Complemento C4b/imunologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Masculino , Dados de Sequência Molecular , Monkeypox virus/genética , Fases de Leitura Aberta/genética , Recombinação Genética , Sciuridae , Carga Viral , Proteínas Virais/genética
14.
J Virol ; 85(6): 2547-56, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191012

RESUMO

Complement activation is an important component of the innate immune response against viral infection and also shapes adaptive immune responses. Despite compelling evidence that complement activation enhances T cell and antibody (Ab) responses during viral infection, it is unknown whether inhibition of complement by pathogens alters these responses. Vaccinia virus (VACV) modulates complement activation by encoding a complement regulatory protein called the vaccinia virus complement control protein (VCP). Although VCP has been described as a virulence factor, the mechanisms by which VCP enhances VACV pathogenesis have not been fully defined. Since complement is necessary for optimal adaptive immune responses to several viruses, we hypothesized that VCP contributes to pathogenesis by modulating anti-VACV T cell and Ab responses. In this study, we used an intradermal model of VACV infection to compare pathogenesis of wild-type virus (vv-VCPwt) and a virus lacking VCP (vv-VCPko). vv-VCPko formed smaller lesions in wild-type mice but not in complement-deficient mice. Attenuation of vv-VCPko correlated with increased accumulation of T cells at the site of infection, enhanced neutralizing antibody responses, and reduced viral titers. Importantly, depleting CD8(+) T cells together with CD4(+) T cells, which also eliminated T helper cell-dependent Ab responses, restored vv-VCPko to wild-type levels of virulence. These results suggest that VCP contributes to virulence by dampening both antibody and T cell responses. This work provides insight into how modulation of complement by poxviruses contributes to virulence and demonstrates that a pathogen-encoded complement regulatory protein can modulate adaptive immunity.


Assuntos
Vaccinia virus/imunologia , Vaccinia virus/patogenicidade , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Anticorpos Neutralizantes/análise , Anticorpos Antivirais/análise , Modelos Animais de Doenças , Feminino , Deleção de Genes , Camundongos , Pele/patologia , Pele/virologia , Linfócitos T/imunologia , Carga Viral , Proteínas Virais/genética , Fatores de Virulência/genética
15.
J Virol ; 84(21): 11245-54, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20719953

RESUMO

The vaccinia virus (VACV) complement control protein (VCP) is an immunomodulatory protein that is both secreted from and expressed on the surface of infected cells. Surface expression of VCP occurs though an interaction with the viral transmembrane protein A56 and is dependent on a free N-terminal cysteine of VCP. Although A56 and VCP have been shown to interact in infected cells, the mechanism remains unclear. To investigate if A56 is sufficient for surface expression, we transiently expressed VCP and A56 in eukaryotic cell lines and found that they interact on the cell surface in the absence of other viral proteins. Since A56 contains three extracellular cysteines, we hypothesized that one of the cysteines may be unpaired and could therefore form a disulfide bridge with VCP. To test this, we generated a series of A56 mutants in which each cysteine was mutated to a serine, and we found that mutation of cysteine 162 abrogated VCP cell surface expression. We also tested the ability of other poxvirus complement control proteins to bind to VACV A56. While the smallpox homolog of VCP is able to bind VACV A56, the ectromelia virus (ECTV) VCP homolog is only able to bind the ECTV homolog of A56, indicating that these proteins may have coevolved. Surface expression of poxvirus complement control proteins may have important implications in viral pathogenesis, as a virus that does not express cell surface VCP is attenuated in vivo. This suggests that surface expression of VCP may contribute to poxvirus pathogenesis.


Assuntos
Membrana Celular/virologia , Poxviridae/patogenicidade , Proteínas Virais/metabolismo , Linhagem Celular , Cisteína/genética , Cisteína/metabolismo , Dissulfetos , Humanos , Mutagênese Sítio-Dirigida
16.
J Virol ; 82(9): 4205-14, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18287241

RESUMO

The vaccinia virus (VACV) complement control protein (VCP) is the major protein secreted from VACV-infected cells. It has been reported that VCP binds to the surfaces of uninfected cells by interacting with heparan sulfate proteoglycans (HSPGs). In this study, we show that VCP is also expressed on the surfaces of infected cells and demonstrate that surface localization occurs independently of HSPGs. Since VCP does not contain a transmembrane domain, we hypothesized that VCP interacts with a membrane protein that localizes to the infected-cell surface. We show that the VACV A56 membrane protein is necessary for the cell surface expression of VCP and demonstrate that VCP and A56 interact in VACV-infected cells. Since the surface expression of VCP was abrogated by reducing agents, we examined the contribution of an unpaired cysteine residue on VCP to VCP surface expression and VCP's interaction with A56. To do this, we mutated the unpaired cysteine in VCP and generated a recombinant virus expressing the altered form of VCP. Following the infection of cells with the mutant virus, VCP was neither expressed on the cell surface nor able to interact with A56. Importantly, the cell surface expression of VCP was found to protect infected cells from complement-mediated lysis. Our findings suggest a new function for VCP that may be important for poxvirus pathogenesis and impact immune responses to VACV-based vaccines.


Assuntos
Ativação do Complemento , Vaccinia virus/patogenicidade , Vacínia/imunologia , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Proteínas Virais/fisiologia , Linhagem Celular , Cisteína/genética , Regulação Viral da Expressão Gênica/imunologia , Heparina/análogos & derivados , Humanos , Mutagênese Sítio-Dirigida , Proteoglicanas , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...