Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 18(8)2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28777313

RESUMO

Arthropod-borne virus (arbovirus) infections cause several emerging and resurgent infectious diseases in humans and animals. Chikungunya-affected areas often overlap with dengue-endemic areas. Concurrent dengue virus (DENV) and chikungunya virus (CHIKV) infections have been detected in travelers returning from regions of endemicity. CHIKV and DENV co-infected Aedes albopictus have also been collected in the vicinity of co-infected human cases, emphasizing the need to study co-infections in mosquitoes. We thus aimed to study the pathogen-pathogen interaction involved in these co-infections in DENV/CHIKV co-infected Aedes aegypti mosquitoes. In mono-infections, we detected CHIKV antigens as early as 4 days post-virus exposure in both the midgut (MG) and salivary gland (SG), whereas we detected DENV serotype 2 (DENV-2) antigens from day 5 post-virus exposure in MG and day 10 post-virus exposure in SG. Identical infection rates were observed for singly and co-infected mosquitoes, and facilitation of the replication of both viruses at various times post-viral exposure. We observed a higher replication for DENV-2 in SG of co-infected mosquitoes. We showed that mixed CHIKV and DENV infection facilitated viral replication in Ae. aegypti. The outcome of these mixed infections must be further studied to increase our understanding of pathogen-pathogen interactions in host cells.


Assuntos
Aedes/virologia , Vírus Chikungunya/fisiologia , Coinfecção/virologia , Vírus da Dengue/fisiologia , Sistema Digestório/virologia , Glândulas Salivares/virologia , Replicação Viral , Administração Oral , Animais , Antígenos Virais/metabolismo , Feminino , Imunofluorescência , Humanos , Imuno-Histoquímica , Recém-Nascido , Cinética , Masculino , RNA Viral/metabolismo , Sorogrupo
2.
J Infect Dis ; 214(2): 281-7, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26977051

RESUMO

The lung is the terminal target of Bacillus anthracis before death, whatever the route of infection (cutaneous, inhalational, or digestive). During a cutaneous infection in absence of toxins, we observed encapsulated bacteria colonizing the alveolar capillary network, bacteria and hemorrhages in alveolar and bronchiolar spaces, and hypoxic foci in the lung (endothelial cells) and brain (neurons and neuropil). Circulating encapsulated bacteria were as chains of approximately 13 µm in length. Bacteria of such size were immediately trapped within the lung capillary network, but bacteria of shorter length were not. Controlling lung-targeted pathology would be beneficial for anthrax treatment.


Assuntos
Antraz/microbiologia , Antraz/patologia , Bacillus anthracis/isolamento & purificação , Capilares/microbiologia , Pulmão/microbiologia , Animais , Capilares/patologia , Modelos Animais de Doenças , Histocitoquímica , Imuno-Histoquímica , Pulmão/patologia , Camundongos , Microscopia Eletrônica de Transmissão
3.
PLoS One ; 7(12): e50464, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23272060

RESUMO

BACKGROUND: Anopheles gambiae is a major vector of malaria and lymphatic filariasis. The arthropod-host interactions occurring at the skin interface are complex and dynamic. We used a global approach to describe the interaction between the mosquito (infected or uninfected) and the skin of mammals during blood feeding. METHODS: Intravital video microscopy was used to characterize several features during blood feeding. The deposition and movement of Plasmodium berghei sporozoites in the dermis were also observed. We also used histological techniques to analyze the impact of infected and uninfected feedings on the skin cell response in naive mice. RESULTS: The mouthparts were highly mobile within the skin during the probing phase. Probing time increased with mosquito age, with possible effects on pathogen transmission. Repletion was achieved by capillary feeding. The presence of sporozoites in the salivary glands modified the behavior of the mosquitoes, with infected females tending to probe more than uninfected females (86% versus 44%). A white area around the tip of the proboscis was observed when the mosquitoes fed on blood from the vessels of mice immunized with saliva. Mosquito feedings elicited an acute inflammatory response in naive mice that peaked three hours after the bite. Polynuclear and mast cells were associated with saliva deposits. We describe the first visualization of saliva in the skin by immunohistochemistry (IHC) with antibodies directed against saliva. Both saliva deposits and sporozoites were detected in the skin for up to 18 h after the bite. CONCLUSION: This study, in which we visualized the probing and engorgement phases of Anopheles gambiae blood meals, provides precise information about the behavior of the insect as a function of its infection status and the presence or absence of anti-saliva antibodies. It also provides insight into the possible consequences of the inflammatory reaction for blood feeding and pathogen transmission.


Assuntos
Anopheles/imunologia , Plasmodium berghei/metabolismo , Animais , Sangue/metabolismo , Culicidae/imunologia , Derme/metabolismo , Feminino , Proteínas de Fluorescência Verde/metabolismo , Interações Hospedeiro-Parasita , Cinética , Malária/transmissão , Camundongos , Microscopia de Fluorescência/métodos , Microscopia de Vídeo/métodos , Modelos Biológicos , Glândulas Salivares/imunologia , Fatores de Tempo
4.
Blood ; 119(11): 2533-44, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22138510

RESUMO

IgE and IgE receptors (FcεRI) are well-known inducers of allergy. We recently found in mice that active systemic anaphylaxis depends on IgG and IgG receptors (FcγRIIIA and FcγRIV) expressed by neutrophils, rather than on IgE and FcεRI expressed by mast cells and basophils. In humans, neutrophils, mast cells, basophils, and eosinophils do not express FcγRIIIA or FcγRIV, but FcγRIIA. We therefore investigated the possible role of FcγRIIA in allergy by generating novel FcγRIIA-transgenic mice, in which various models of allergic reactions induced by IgG could be studied. In mice, FcγRIIA was sufficient to trigger active and passive anaphylaxis, and airway inflammation in vivo. Blocking FcγRIIA in vivo abolished these reactions. We identified mast cells to be responsible for FcγRIIA-dependent passive cutaneous anaphylaxis, and monocytes/macrophages and neutrophils to be responsible for FcγRIIA-dependent passive systemic anaphylaxis. Supporting these findings, human mast cells, monocytes and neutrophils produced anaphylactogenic mediators after FcγRIIA engagement. IgG and FcγRIIA may therefore contribute to allergic and anaphylactic reactions in humans.


Assuntos
Hipersensibilidade/etiologia , Hipersensibilidade/patologia , Inflamação/etiologia , Inflamação/patologia , Anafilaxia Cutânea Passiva , Receptores de IgG/fisiologia , Sistema Respiratório/patologia , Animais , Células Cultivadas , Eosinófilos/imunologia , Eosinófilos/metabolismo , Eosinófilos/patologia , Humanos , Hipersensibilidade/metabolismo , Inflamação/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Sistema Respiratório/metabolismo
5.
J Infect Dis ; 205(1): 134-43, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22090450

RESUMO

BACKGROUND: Although laboratory mice are usually highly susceptible to Yersinia pestis, we recently identified a mouse strain (SEG) that exhibited an exceptional capacity to resist bubonic plague and used it to identify immune mechanisms associated with resistance. METHODS: The kinetics of infection, circulating blood cells, granulopoiesis, lesions, and cellular populations in the spleen, and cytokine production in various tissues were compared in SEG and susceptible C57BL/6J mice after subcutaneous infection with the virulent Y. pestis CO92. RESULTS: Bacterial invasion occurred early (day 2) but was transient in SEG/Pas mice, whereas in C57BL/6J mice it was delayed but continuous until death. The bacterial load in all organs significantly correlated with the production of 5 cytokines (granulocyte colony-stimulating factor, keratinocyte-derived chemokine (KC), macrophage cationic peptide-1 (MCP-1), interleukin 1α, and interleukin 6) involved in monocyte and neutrophil recruitment. Indeed, higher proportions of these 2 cell types in blood and massive recruitment of F4/80(+)CD11b(-) macrophages in the spleen were observed in SEG/Pas mice at an early time point (day 2). Later times after infection (day 4) were characterized in C57BL/6J mice by destructive lesions of the spleen and impaired granulopoiesis. CONCLUSION: A fast and efficient Y. pestis dissemination in SEG mice may be critical for the triggering of an early and effective innate immune response necessary for surviving plague.


Assuntos
Citocinas/metabolismo , Imunidade Inata , Camundongos Endogâmicos/imunologia , Peste/imunologia , Yersinia pestis/patogenicidade , Animais , Carga Bacteriana , Quimiocinas/metabolismo , Resistência à Doença , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos/metabolismo , Fagócitos/imunologia , Peste/metabolismo , Peste/microbiologia , Yersinia pestis/imunologia
6.
Am J Pathol ; 178(6): 2523-35, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21641378

RESUMO

Powerful noninvasive imaging technologies enable real-time tracking of pathogen-host interactions in vivo, giving access to previously elusive events. We visualized the interactions between wild-type Bacillus anthracis and its host during a spore infection through bioluminescence imaging coupled with histology. We show that edema toxin plays a central role in virulence in guinea pigs and during inhalational infection in mice. Edema toxin (ET), but not lethal toxin (LT), markedly modified the patterns of bacterial dissemination leading, to apparent direct dissemination to the spleen and provoking apoptosis of lymphoid cells. Each toxin alone provoked particular histological lesions in the spleen. When ET and LT are produced together during infection, a specific temporal pattern of lesion developed, with early lesions typical of LT, followed at a later stage by lesions typical of ET. Our study provides new insights into the complex spatial and temporal effects of B. anthracis toxins in the infected host, suggesting a greater role than previously suspected for ET in anthrax and suggesting that therapeutic targeting of ET contributes to protection.


Assuntos
Antraz/microbiologia , Antraz/patologia , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Diagnóstico por Imagem/métodos , Fatores de Virulência/imunologia , Animais , Antraz/prevenção & controle , Apoptose , Bacillus anthracis/patogenicidade , Feminino , Cobaias/microbiologia , Exposição por Inalação , Luminescência , Camundongos , Camundongos Endogâmicos BALB C , Nasofaringe/microbiologia , Nasofaringe/patologia , Testes de Neutralização , Pele/microbiologia , Pele/patologia , Baço/microbiologia , Baço/patologia , Fatores de Tempo
7.
J Immunol ; 186(4): 1899-903, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21248252

RESUMO

K/BxN serum-induced passive arthritis was reported to depend on the activation of mast cells, triggered by the activating IgG receptor FcγRIIIA, when engaged by IgG1 autoantibodies present in K/BxN serum. This view is challenged by the fact that FcγRIIIA-deficient mice still develop K/BxN arthritis and because FcγRIIIA is the only activating IgG receptor expressed by mast cells. We investigated the contribution of IgG receptors, IgG subclasses, and cells in K/BxN arthritis. We found that the activating IgG2 receptor FcγRIV, expressed only by monocytes/macrophages and neutrophils, was sufficient to induce disease. K/BxN arthritis occurred not only in mast cell-deficient W(sh) mice, but also in mice whose mast cells express no activating IgG receptors. We propose that at least two autoantibody isotypes, IgG1 and IgG2, and two activating IgG receptors, FcγRIIIA and FcγRIV, contribute to K/BxN arthritis, which requires at least two cell types other than mast cells, monocytes/macrophages, and neutrophils.


Assuntos
Artrite Experimental/imunologia , Autoanticorpos/fisiologia , Sítios de Ligação de Anticorpos , Receptores de IgG/fisiologia , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Autoanticorpos/sangue , Imunoglobulina G/metabolismo , Mastócitos/imunologia , Mastócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Receptores de IgG/deficiência , Receptores de IgG/genética , Soro/fisiologia
8.
J Exp Med ; 207(11): 2313-22, 2010 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-20956545

RESUMO

Streptococcus agalactiae (group B streptococcus; GBS) is a normal constituent of the intestinal microflora and the major cause of human neonatal meningitis. A single clone, GBS ST-17, is strongly associated with a deadly form of the infection called late-onset disease (LOD), which is characterized by meningitis in infants after the first week of life. The pathophysiology of LOD remains poorly understood, but our epidemiological and histopathological results point to an oral route of infection. Here, we identify a novel ST-17-specific surface-anchored protein that we call hypervirulent GBS adhesin (HvgA), and demonstrate that its expression is required for GBS hypervirulence. GBS strains that express HvgA adhered more efficiently to intestinal epithelial cells, choroid plexus epithelial cells, and microvascular endothelial cells that constitute the blood-brain barrier (BBB), than did strains that do not express HvgA. Heterologous expression of HvgA in nonadhesive bacteria conferred the ability to adhere to intestinal barrier and BBB-constituting cells. In orally inoculated mice, HvgA was required for intestinal colonization and translocation across the intestinal barrier and the BBB, leading to meningitis. In conclusion, HvgA is a critical virulence trait of GBS in the neonatal context and stands as a promising target for the development of novel diagnostic and antibacterial strategies.


Assuntos
Adesinas Bacterianas/metabolismo , Translocação Bacteriana/fisiologia , Barreira Hematoencefálica/metabolismo , Mucosa Intestinal/metabolismo , Meninges/metabolismo , Meningites Bacterianas/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus agalactiae , Adesinas Bacterianas/genética , Animais , Aderência Bacteriana/fisiologia , Barreira Hematoencefálica/microbiologia , Feminino , Células HeLa , Humanos , Lactente , Recém-Nascido , Intestinos/microbiologia , Masculino , Meninges/microbiologia , Meningites Bacterianas/genética , Meningites Bacterianas/microbiologia , Camundongos , Especificidade de Órgãos , Infecções Estreptocócicas/genética , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/patogenicidade , Streptococcus agalactiae/fisiologia
9.
PLoS One ; 5(10)2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-20957153

RESUMO

BACKGROUND: Arthropod borne virus infections cause several emerging and resurgent infectious diseases. Among the diseases caused by arboviruses, dengue and chikungunya are responsible for a high rate of severe human diseases worldwide. The midgut of mosquitoes is the first barrier for pathogen transmission and is a target organ where arboviruses must replicate prior to infecting other organs. A proteomic approach was undertaken to characterize the key virus/vector interactions and host protein modifications that happen in the midgut for viral transmission to eventually take place. METHODOLOGY AND PRINCIPAL FINDINGS: Using a proteomics differential approach with two-Dimensional Differential in-Gel Electrophoresis (2D-DIGE), we defined the protein modulations in the midgut of Aedes aegypti that were triggered seven days after an oral infection (7 DPI) with dengue 2 (DENV-2) and chikungunya (CHIKV) viruses. Gel profile comparisons showed that the level of 18 proteins was modulated by DENV-2 only and 12 proteins were modulated by CHIKV only. Twenty proteins were regulated by both viruses in either similar or different ways. Both viruses caused an increase of proteins involved in the generation of reactive oxygen species, energy production, and carbohydrate and lipid metabolism. Midgut infection by DENV-2 and CHIKV triggered an antioxidant response. CHIKV infection produced an increase of proteins involved in detoxification. CONCLUSION/SIGNIFICANCE: Our study constitutes the first analysis of the protein response of Aedes aegypti's midgut infected with viruses belonging to different families. It shows that the differentially regulated proteins in response to viral infection include structural, redox, regulatory proteins, and enzymes for several metabolic pathways. Some of these proteins like antioxidant are probably involved in cell protection. On the other hand, we propose that the modulation of other proteins like transferrin, hsp60 and alpha glucosidase, may favour virus survival, replication and transmission, suggesting a subversion of the insect cell metabolism by the arboviruses.


Assuntos
Aedes/metabolismo , Vírus Chikungunya/isolamento & purificação , Vírus da Dengue/isolamento & purificação , Proteínas de Insetos/metabolismo , Mucosa Intestinal/metabolismo , Aedes/virologia , Animais , Metabolismo dos Carboidratos , Eletroforese em Gel Bidimensional , Metabolismo Energético , Imunofluorescência , Metabolismo dos Lipídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
10.
J Infect Dis ; 200(9): 1381-9, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19785525

RESUMO

BACKGROUND: The Bacillus anthracis poly-gamma-D-glutamate capsule is essential for virulence. It impedes phagocytosis and protects bacilli from the immune system, thus promoting systemic dissemination. METHODS: To further define the virulence mechanisms brought into play by the capsule, we characterized the interactions between encapsulated nontoxinogenic B. anthracis and its host in vivo through histological analysis, perfusion, and competition experiments with purified capsule. RESULTS: Clearance of encapsulated bacilli from the blood was rapid (>90% clearance within 5 min), with 75% of the bacteria being trapped in the liver. Competition experiments with purified capsule polyglutamate inhibited this interaction. At the septicemic phase of cutaneous infection with spores, the encapsulated bacilli were trapped in the vascular spaces of the liver and interacted closely with the liver endothelium in the sinusoids and terminal and portal veins. They often grow as microcolonies containing capsular material shed by the bacteria. CONCLUSION: We show that, in addition to its inhibitory effect on the interaction with the immune system, the capsule surrounding B. anthracis plays an active role in mediating the trapping of the bacteria within the liver and may thus contribute to anthrax pathogenesis. Because other microorganisms produce polyglutamate, it may also represent a general mechanism of virulence or in vivo survival.


Assuntos
Antraz/microbiologia , Bacillus anthracis/patogenicidade , Cápsulas Bacterianas/fisiologia , Endotélio/microbiologia , Fígado/microbiologia , Animais , Feminino , Interações Hospedeiro-Patógeno , Camundongos , Ácido Poliglutâmico/fisiologia , Virulência
11.
J Immunol ; 183(1): 670-6, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535624

RESUMO

Burkholderia cenocepacia is an opportunistic pathogen of major concern for cystic fibrosis patients as well as immunocompromised cancer patients and transplant recipients. The mechanisms by which B. cenocepacia triggers a rapid health deterioration of the susceptible host have yet to be characterized. TLR and their key signaling intermediate MyD88 play a central role in the detection of microbial molecular patterns and in the initiation of an effective immune response. We performed a study to better understand the role of TLR-MyD88 signaling in B. cenocepacia-induced pathogenesis in the immunocompromised host, using an experimental murine model. The time-course of several dynamic parameters, including animal survival, bacterial load, and secretion of critical inflammatory mediators, was compared in infected and immunosuppressed wild-type and MyD88(-/-) mice. Notably, when compared with wild-type mice, infected MyD88(-/-) animals displayed significantly reduced levels of inflammatory mediators (including KC, TNF-alpha, IL-6, MIP-2, and G-CSF) in blood and lung airspaces. Moreover, despite a higher transient bacterial load in the lungs, immunosuppressed mice deficient in MyD88 had an unexpected survival advantage. Finally, we showed that this B. cenocepacia-induced life-threatening infection of wild-type mice involved the proinflammatory cytokine TNF-alpha and could be prevented by corticosteroids. Altogether, our findings demonstrate that a MyD88-dependent pathway can critically contribute to a detrimental host inflammatory response that leads to fatal pneumonia.


Assuntos
Infecções por Burkholderia/prevenção & controle , Burkholderia cepacia , Hospedeiro Imunocomprometido/genética , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Infecções Oportunistas/prevenção & controle , Pneumonia Bacteriana/prevenção & controle , Animais , Infecções por Burkholderia/imunologia , Infecções por Burkholderia/mortalidade , Burkholderia cepacia/crescimento & desenvolvimento , Burkholderia cepacia/imunologia , Burkholderia cepacia/patogenicidade , Hospedeiro Imunocomprometido/imunologia , Imunossupressores/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , Infecções Oportunistas/imunologia , Infecções Oportunistas/mortalidade , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/mortalidade , Análise de Sobrevida , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia
12.
Blood ; 112(6): 2520-8, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18579796

RESUMO

The current paradigm in Plasmodium falciparum malaria pathogenesis states that young, ring-infected erythrocytes (rings) circulate in peripheral blood and that mature stages are sequestered in the vasculature, avoiding clearance by the spleen. Through ex vivo perfusion of human spleens, we examined the interaction of this unique blood-filtering organ with P falciparum-infected erythrocytes. As predicted, mature stages were retained. However, more than 50% of rings were also retained and accumulated upstream from endothelial sinus wall slits of the open, slow red pulp microcirculation. Ten percent of rings were retained at each spleen passage, a rate matching the proportion of blood flowing through the slow circulatory compartment established in parallel using spleen contrast-enhanced ultrasonography in healthy volunteers. Rings displayed a mildly but significantly reduced elongation index, consistent with a retention process, due to their altered mechanical properties. This raises the new paradigm of a heterogeneous ring population, the less deformable subset being retained in the spleen, thereby reducing the parasite biomass that will sequester in vital organs, influencing the risk of severe complications, such as cerebral malaria or severe anemia. Cryptic ring retention uncovers a new role for the spleen in the control of parasite density, opening novel intervention opportunities.


Assuntos
Eritrócitos/parasitologia , Microcirculação/parasitologia , Plasmodium falciparum , Baço/irrigação sanguínea , Animais , Velocidade do Fluxo Sanguíneo , Humanos , Técnicas In Vitro , Perfusão , Fluxo Sanguíneo Regional , Baço/parasitologia
13.
PLoS One ; 2(11): e1168, 2007 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18000540

RESUMO

BACKGROUND: A Chikungunya (CHIK) outbreak hit La Réunion Island in 2005-2006. The implicated vector was Aedes albopictus. Here, we present the first study on the susceptibility of Ae. albopictus populations to sympatric CHIKV isolates from La Réunion Island and compare it to other virus/vector combinations. METHODOLOGY AND FINDINGS: We orally infected 8 Ae. albopictus collections from La Réunion and 3 from Mayotte collected in March 2006 with two Chikungunya virus (CHIKV) from La Réunion: (i) strain 05.115 collected in June 2005 with an Alanine at the position 226 of the glycoprotein E1 and (ii) strain 06.21 collected in November 2005 with a substitution A226V. Two other CHIKV isolates and four additional mosquito strains/species were also tested. The viral titer of the infectious blood-meal was 10(7) plaque forming units (pfu)/mL. Dissemination rates were assessed by immunofluorescent staining on head squashes of surviving females 14 days after infection. Rates were at least two times higher with CHIKV 06.21 compared to CHIKV 05.115. In addition, 10 individuals were analyzed every day by quantitative RT-PCR. Viral RNA was quantified on (i) whole females and (ii) midguts and salivary glands of infected females. When comparing profiles, CHIKV 06.21 produced nearly 2 log more viral RNA copies than CHIKV 05.115. Furthermore, females infected with CHIKV 05.115 could be divided in two categories: weakly susceptible or strongly susceptible, comparable to those infected by CHIKV 06.21. Histological analysis detected the presence of CHIKV in salivary glands two days after infection. In addition, Ae. albopictus from La Réunion was as efficient vector as Ae. aegypti and Ae. albopictus from Vietnam when infected with the CHIKV 06.21. CONCLUSIONS: Our findings support the hypothesis that the CHIK outbreak in La Réunion Island was due to a highly competent vector Ae. albopictus which allowed an efficient replication and dissemination of CHIKV 06.21.


Assuntos
Aedes/virologia , Infecções por Alphavirus/epidemiologia , Vírus Chikungunya/isolamento & purificação , Infecções por Alphavirus/virologia , Animais , Vírus Chikungunya/classificação , Surtos de Doenças , Feminino , Masculino , Filogenia
14.
Am J Pathol ; 170(3): 1003-17, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322384

RESUMO

When intraperitoneally injected into Swiss mice, Clostridium sordellii lethal toxin reproduces the fatal toxic shock syndrome observed in humans and animals after natural infection. This animal model was used to study the mechanism of lethal toxin-induced death. Histopathological and biochemical analyses identified lung and heart as preferential organs targeted by lethal toxin. Massive extravasation of blood fluid in the thoracic cage, resulting from an increase in lung vascular permeability, generated profound modifications such as animal dehydration, increase in hematocrit, hypoxia, and finally, cardiorespiratory failure. Vascular permeability increase induced by lethal toxin resulted from modifications of lung endothelial cells as evidenced by electron microscopy. Immunohistochemical analysis demonstrated that VE-cadherin, a protein participating in intercellular adherens junctions, was redistributed from membrane to cytosol in lung endothelial cells. No major sign of lethal toxin-induced inflammation was observed that could participate in the toxic shock syndrome. The main effect of the lethal toxin is the glucosylation-dependent inactivation of small GTPases, in particular Rac, which is involved in actin polymerization occurring in vivo in lungs leading to E-cadherin junction destabilization. We conclude that the cells most susceptible to lethal toxin are lung vascular endothelial cells, the adherens junctions of which were altered after intoxication.


Assuntos
Toxinas Bacterianas/toxicidade , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Edema Pulmonar/induzido quimicamente , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/patologia , Animais , Caderinas/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Modelos Animais de Doenças , Edema Cardíaco/induzido quimicamente , Edema Cardíaco/patologia , Células Endoteliais/patologia , Coração/efeitos dos fármacos , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Camundongos , Microscopia Eletrônica de Transmissão , Edema Pulmonar/patologia , Choque Séptico/induzido quimicamente , Proteínas rac de Ligação ao GTP/efeitos dos fármacos
15.
Blood ; 107(9): 3745-52, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16384927

RESUMO

The spleen plays a central role in the pathophysiology of several potentially severe diseases such as inherited red cell membrane disorders, hemolytic anemias, and malaria. Research on these diseases is hampered by ethical constraints that limit human spleen tissue explorations. We identified a surgical situation--left splenopancreatectomy for benign pancreas tumors--allowing spleen retrieval at no risk for patients. Ex vivo perfusion of retrieved intact spleens for 4 to 6 hours maintained a preserved parenchymal structure, vascular flow, and metabolic activity. Function preservation was assessed by testing the ability of isolated-perfused spleens to retain Plasmodium falciparum-infected erythrocytes preexposed to the antimalarial drug artesunate (Art-iRBCs). More than 95% of Art-iRBCs were cleared from the perfusate in 2 hours. At each transit through isolated-perfused spleens, parasite remnants were removed from 0.2% to 0.23% of Art-iRBCs, a proportion consistent with the 0.02% to 1% pitting rate previously established in artesunate-treated patients. Histologic analysis showed that more than 90% of Art-iRBCs were retained and processed in the red pulp, providing the first direct evidence of a zone-dependent parasite clearance by the human spleen. Human-specific physiologic or pathophysiologic mechanisms involving clearing or processing functions of the spleen can now be experimentally explored in a human tissue context.


Assuntos
Baço/fisiologia , Animais , Antimaláricos/farmacologia , Artemisininas/farmacologia , Artesunato , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Humanos , Técnicas In Vitro , Malária/parasitologia , Malária/fisiopatologia , Perfusão , Fagocitose , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/patogenicidade , Sesquiterpenos/farmacologia , Baço/irrigação sanguínea , Baço/imunologia , Baço/parasitologia
16.
J Immunol ; 174(6): 3570-9, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15749894

RESUMO

The chromosomal locus encoding the early secreted antigenic target, 6 kDa (ESAT-6) secretion system 1 of Mycobacterium tuberculosis, also referred to as "region of difference 1 (RD1)," is absent from Mycobacterium bovis bacillus Calmette-Guerin (BCG). In this study, using low-dose aerosol infection in mice, we demonstrate that BCG complemented with RD1 (BCG::RD1) displays markedly increased virulence which albeit does not attain that of M. tuberculosis H37Rv. Nevertheless, phenotypic and functional analyses of immune cells at the site of infection show that the capacity of BCG::RD1 to initiate recruitment/activation of immune cells is comparable to that of fully virulent H37Rv. Indeed, in contrast to the parental BCG, BCG::RD1 mimics H37Rv and induces substantial influx of activated (CD44highCD45RB(-)CD62L(-)) or effector (CD45RB(-)CD27(-)) T cells and of activated CD11c(+)CD11bhigh cells to the lungs of aerosol-infected mice. For the first time, using in vivo analysis of transcriptome of inflammatory cytokines and chemokines of lung interstitial CD11c+ cells, we show that in a low-dose aerosol infection model, BCG::RD1 triggered an activation/inflammation program comparable to that induced by H37Rv while parental BCG, due to its overattenuation, did not initiate the activation program in lung interstitial CD11c+ cells. Thus, products encoded by the ESAT-6 secretion system 1 of M. tuberculosis profoundly modify the interaction between mycobacteria and the host innate and adaptive immune system. These modifications can explain the previously described improved protective capacity of BCG::RD1 vaccine candidate against M. tuberculosis challenge.


Assuntos
Antígenos de Bactérias/imunologia , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Animais , Antígenos de Bactérias/genética , Vacina BCG/imunologia , Vacina BCG/farmacologia , Proteínas de Bactérias , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Quimiocinas/genética , Citocinas/genética , Células Dendríticas/imunologia , Expressão Gênica , Teste de Complementação Genética , Imunidade Inata , Técnicas In Vitro , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium bovis/genética , Mycobacterium bovis/imunologia , Mycobacterium bovis/patogenicidade , Mycobacterium tuberculosis/genética , Receptores de Citocinas/genética , Tuberculose Pulmonar/etiologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/prevenção & controle , Virulência/genética , Virulência/imunologia
17.
Infect Immun ; 73(3): 1771-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15731078

RESUMO

The protozoan parasite Entamoeba histolytica colonizes the human large bowel. Invasion of the intestinal epithelium causes amoebic colitis and opens the route for amoebic liver abscesses. The parasite relies on its dynamic actomyosin cytoskeleton and on surface adhesion molecules for dissemination in the human tissues. Here we show that the galactose/N-acetylgalactosamine (Gal/GalNAc) lectin clusters in focal structures localized in the region of E. histolytica that contacts monolayers of enterocytes. Disruption of myosin II activity impairs the formation of these structures and renders the trophozoites avirulent for liver abscess development. Production of the cytoplasmic domain of the E. histolytica Gal/GalNAc lectin in engineered trophozoites causes reduced adhesion to enterocytes. Intraportal delivery of these parasites to the liver leads to the formation of a large number of small abscesses with disorganized morphology that are localized in the vicinity of blood vessels. The data support a model for invasion in which parasite motility is essential for establishment of infectious foci, while the adhesion to host cells modulates the distribution of trophozoites in the liver and their capacity to migrate in the hepatic tissue.


Assuntos
Citoesqueleto/metabolismo , Entamoeba histolytica/fisiologia , Entamoeba histolytica/patogenicidade , Entamebíase/fisiopatologia , Abscesso Hepático Amebiano/fisiopatologia , Animais , Adesão Celular , Cricetinae , Entamebíase/parasitologia , Entamebíase/patologia , Enterócitos/parasitologia , Humanos , Lectinas/metabolismo , Abscesso Hepático Amebiano/parasitologia , Abscesso Hepático Amebiano/patologia , Masculino , Mesocricetus , Miosinas/metabolismo , Virulência
18.
Proc Natl Acad Sci U S A ; 101(16): 6152-7, 2004 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-15073336

RESUMO

Listeria monocytogenes produces severe fetoplacental infections in humans. How it targets and crosses the maternofetal barrier is unknown. We used immunohistochemistry to examine the location of L. monocytogenes in placental and amniotic tissue samples obtained from women with fetoplacental listeriosis. The results raised the possibility that L. monocytogenes crosses the maternofetal barrier through the villous syncytiotrophoblast, with secondary infection occurring via the amniotic epithelium. Because epidemiological studies indicate that the bacterial surface protein, internalin (InlA), may play a role in human fetoplacental listeriosis, we investigated the cellular patterns of expression of its host receptor, E-cadherin, at the maternofetal interface. E-cadherin was found on the basal and apical plasma membranes of syncytiotrophoblasts and in villous cytotrophoblasts. Established trophoblastic cell lines, primary trophoblast cultures, and placental villous explants were each exposed to isogenic InlA+ or InlA- strains of L. monocytogenes, and to L. innocua expressing or not InlA. Quantitative assays of cellular invasion demonstrated that bacterial entry into syncytiotrophoblasts occurs via the apical membrane in an InlA-E-cadherin dependent manner. In human placental villous explants, bacterial invasion of the syncytiotrophoblast barrier and underlying villous tissue and subsequent replication produces histopathological lesions that mimic those seen in placentas of women with listeriosis. Thus, the InlA-E-cadherin interaction that plays a key role in the crossing of the intestinal barrier in humans is also exploited by L. monocytogenes to target and cross the placental barrier. Such a ligand-receptor interaction allowing a pathogen to specifically cross the placental villous trophoblast barrier has not been reported previously.


Assuntos
Proteínas de Bactérias/fisiologia , Caderinas/metabolismo , Listeria monocytogenes/fisiologia , Listeriose/transmissão , Troca Materno-Fetal , Complicações Infecciosas na Gravidez , Trofoblastos/metabolismo , Feminino , Humanos , Placenta/metabolismo , Placenta/microbiologia , Gravidez , Ligação Proteica
19.
J Bone Miner Res ; 19(1): 100-10, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14753742

RESUMO

UNLABELLED: The pluripotent mesoblastic C1 cell line was used under serum-free culture conditions to investigate how paracrine and autocrine signals cooperate to drive chondrogenesis. Sequential addition of two systemic hormones, dexamethasone and triiodothyronine, permits full chondrogenic differentiation. The cell intrinsic activation of the BMP signaling pathway and Sox9 expression occurring on mesoblastic condensation is insufficient for recruitment of the progenitors. Dexamethasone-dependent Sox9 upregulation is essential for chondrogenesis. INTRODUCTION: Differentiation of lineage stem cells relies on cell autonomous regulations modulated by external signals. We used the pluripotent mesoblastic C1 cell line under serum-free culture conditions to investigate how paracrine and autocrine signals cooperate to induce differentiation of a precursor clone along the chondrogenic lineage. MATERIALS AND METHODS: C1 cells, cultured as aggregates, were induced toward chondrogenesis by addition of 10(-7) M dexamethasone in serum-free medium. After 30 days, dexamethasone was replaced by 10 nM triiodothyronine to promote final hypertrophic conversion. Mature and hypertrophic phenotypes were characterized by immunocytochemistry using specific antibodies against types II and X collagens, respectively. Type II collagen, bone morphogenetic proteins (BMPs), BMP receptors, Smads, and Sox9 expression were monitored by reverse transcriptase-polymerase chain reaction (RT-PCR), Northern blot, and/or Western blot analysis. RESULTS AND CONCLUSIONS: Once C1 cells have formed nodules, sequential addition of two systemic hormones is sufficient to promote full chondrogenic differentiation. In response to dexamethasone, nearly 100% of the C1 precursors engage in chondrogenesis and convert within 30 days into mature chondrocytes, which triggers a typical cartilage matrix. On day 25, a switch in type II procollagen mRNA splicing acted as a limiting step in the acquisition of the mature chondrocyte phenotype. On day 30, substitution of dexamethasone with triiodothyronine triggers the final differentiation into hypertrophic chondrocytes within a further 15 days. The chondrogenic process is supported by intrinsic expression of Sox9 and BMP family genes. Similarly to the in vivo situation, activation of Sox9 expression and the BMP signaling pathway occurred on mesoblastic condensation. After induction, BMP-activated Smad nuclear translocation persisted throughout the process until the onset of hypertrophy. After dexamethasone addition, Sox9 expression was upregulated. Dexamethasone withdrawal reversed the increase in Sox9 expression and stopped differentiation. Thus, Sox9 seems to be a downstream mediator of dexamethasone action.


Assuntos
Comunicação Autócrina/fisiologia , Diferenciação Celular/fisiologia , Condrócitos/fisiologia , Comunicação Parácrina/fisiologia , Células-Tronco Pluripotentes/fisiologia , Transdução de Sinais/fisiologia , Agrecanas , Animais , Northern Blotting , Receptores de Proteínas Morfogenéticas Ósseas , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Agregação Celular/fisiologia , Linhagem Celular Tumoral , Condrócitos/citologia , Condrócitos/metabolismo , Colágeno Tipo I/análise , Colágeno Tipo I/metabolismo , Colágeno Tipo II/análise , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Colágeno Tipo X/análise , Colágeno Tipo X/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Dexametasona/farmacologia , Proteínas da Matriz Extracelular/análise , Regulação Neoplásica da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/farmacologia , Lectinas Tipo C , Camundongos , Osteopontina , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas/análise , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOX9 , Sialoglicoproteínas/análise , Sialoglicoproteínas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Tri-Iodotironina/farmacologia , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...