Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
PLoS Pathog ; 18(5): e1010511, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35605029

RESUMO

Hematogenous dissemination is a critical step in the evolution of local infection to systemic disease. The Lyme disease (LD) spirochete, which efficiently disseminates to multiple tissues, has provided a model for this process, in particular for the key early event of pathogen adhesion to the host vasculature. This occurs under shear force mediated by interactions between bacterial adhesins and mammalian cell-surface proteins or extracellular matrix (ECM). Using real-time intravital imaging of the Lyme spirochete in living mice, we previously identified BBK32 as the first LD spirochetal adhesin demonstrated to mediate early vascular adhesion in a living mouse; however, deletion of bbk32 resulted in loss of only about half of the early interactions, suggesting the existence of at least one other adhesin (adhesin-X) that promotes early vascular interactions. VlsE, a surface lipoprotein, was identified long ago by its capacity to undergo rapid antigenic variation, is upregulated in the mammalian host and required for persistent infection in immunocompetent mice. In immunodeficient mice, VlsE shares functional overlap with OspC, a multi-functional protein that displays dermatan sulfate-binding activity and is required for joint invasion and colonization. In this research, using biochemical and genetic approaches as well as intravital imaging, we have identified VlsE as adhesin-X; it is a dermatan sulfate (DS) adhesin that efficiently promotes transient adhesion to the microvasculature under shear force via its DS binding pocket. Intravenous inoculation of mice with a low-passage infectious B. burgdorferi strain lacking both bbk32 and vlsE almost completely eliminated transient microvascular interactions. Comparative analysis of binding parameters of VlsE, BBK32 and OspC provides a possible explanation why these three DS adhesins display different functionality in terms of their ability to promote early microvascular interactions.


Assuntos
Adesinas Bacterianas , Variação Antigênica , Antígenos de Bactérias , Proteínas de Bactérias , Borrelia burgdorferi , Lipoproteínas , Doença de Lyme , Microvasos , Adesinas Bacterianas/genética , Adesinas Bacterianas/imunologia , Animais , Variação Antigênica/genética , Variação Antigênica/imunologia , Antígenos de Bactérias/genética , Antígenos de Bactérias/imunologia , Aderência Bacteriana/genética , Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Borrelia burgdorferi/genética , Borrelia burgdorferi/imunologia , Dermatan Sulfato/imunologia , Lipoproteínas/genética , Lipoproteínas/imunologia , Doença de Lyme/genética , Doença de Lyme/imunologia , Doença de Lyme/microbiologia , Mamíferos , Camundongos , Microvasos/imunologia , Microvasos/microbiologia , Resistência ao Cisalhamento
2.
Ticks Tick Borne Dis ; 11(4): 101451, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32360026

RESUMO

Lyme borreliosis is one of the major tick-borne diseases in Europe. Events of the translocation of Borrelia across the blood-brain barrier (BBB) involve multiple interactions between borrelial surface proteins and receptors on the brain microvascular endothelial cells (hBMECs). In this study, we aimed to identify proteins of Borrelia that plausibly interact with hBMECs. The surface proteome of live Borrelia (a neuroinvasive strain of B. garinii) was crosslinked with biotin prior to its incubation with hBMECs. The interacting proteins were recovered by affinity purification, followed by SWATH-MS. Twenty-four interacting candidates were grouped into outer membrane proteins (n = 12) and inner membrane proteins (n = 12) based on the subcellular location as per the predictions of LocateP. Other algorithms like TMHMM 2.0 and LipoP, ontology search and literature review were subsequently applied to each of the identified protein candidates to shortlist the most probable interactors. Six proteins namely, LysM domain protein, BESBP-5, Antigen S1, CRASP-1 (Bg071), Erp23 protein and Mlp family Lipoprotein were selected to produce their recombinant forms and experimentally validate their interaction with hBMECs. All the recombinant proteins interacted with hBMECs, in ELISA and immunocytochemistry. We present here a high-throughput approach of generating a dataset of plausible borrelial ligands followed by a systematic bioinformatic pipeline to categorize the proteins for experimental validation.


Assuntos
Proteínas de Bactérias/genética , Grupo Borrelia Burgdorferi/genética , Encéfalo/microbiologia , Células Endoteliais/microbiologia , Microvasos/microbiologia , Proteoma/metabolismo , Proteínas de Bactérias/metabolismo , Grupo Borrelia Burgdorferi/metabolismo , Doença de Lyme
3.
Cytokine ; 121: 154737, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31158700

RESUMO

Ureaplasma species (spp.) are considered commensals of the adult urogenital tract, but may cause chorioamnionitis and preterm birth as well as sepsis and meningitis in neonates. Pathomechanisms in Ureaplasma-driven neuroinflammation are largely unknown. This study addressed mRNA and protein expression of intercellular and vascular cell adhesion molecules (ICAM-1, VCAM-1), granulocyte-colony stimulating factor (G-CSF), and vascular endothelial growth factor (VEGF) in native or lipopolysaccharide (LPS) co-stimulated human brain microvascular endothelial cells (HBMEC) exposed to Ureaplasma (U.) urealyticum or U. parvum. Ureaplasma spp. reduced G-CSF mRNA (p < 0.05) and protein expression (p < 0.01) and increased VEGF mRNA levels (p < 0.01) in native HBMEC. Upon co-stimulation, Ureaplasma isolates enhanced LPS-evoked VEGF and ICAM-1 mRNA expression (p < 0.05), but mitigated G-CSF and VCAM-1 mRNA responses (p < 0.05). In line with previous findings, our results indicate an ability of Ureaplasma spp. to compromise blood-brain barrier integrity, mitigate immune defense, and subdue neuroprotective mechanisms. This may facilitate intracerebral inflammation, allow chronic infections, and promote brain injury. More pronounced effects in co-stimulated cells may indicate an immunomodulatory capacity of Ureaplasma spp.


Assuntos
Encéfalo/irrigação sanguínea , Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Microvasos/microbiologia , Ureaplasma/fisiologia , Adulto , Encéfalo/patologia , Moléculas de Adesão Celular/genética , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética
4.
Artigo em Inglês | MEDLINE | ID: mdl-29963502

RESUMO

Neurobrucellosis is an inflammatory disease caused by the invasion of Brucella spp. to the central nervous system (CNS). The pathogenesis of the disease is not well characterized; however, for Brucella to gain access to the brain parenchyma, traversing of the blood-brain barrier (BBB) must take place. To understand the CNS determinants of the pathogenesis of B. abortus, we have used the in vitro BBB model of human brain microvascular endothelial cells (HBMEC) to study the interactions between B. abortus and brain endothelial cells. In this study, we showed that B. abortus is able to adhere and invade HBMEC which was dependent on microtubules, microfilaments, endosome acidification and de novo protein synthesis. After infection, B. abortus rapidly escapes the endosomal compartment of HBMEC and forms a replicative Brucella-containing vacuole that involves interactions with the endoplasmic reticulum. Despite the ability of B. abortus to invade and replicate in HBMEC, the bacterium was unable by itself to traverse HBMEC, but could traverse polarized HBMEC monolayers within infected monocytes. Importantly, infected monocytes that traversed the HBMEC monolayer were a bacterial source for de novo infection of glial cells. This is the first demonstration of the mechanism whereby B. abortus is able to traverse the BBB and infect cells of the CNS. These results may have important implications in our understanding of the pathogenesis of neurobrucellosis.


Assuntos
Barreira Hematoencefálica/microbiologia , Brucella abortus/crescimento & desenvolvimento , Células Endoteliais/microbiologia , Leucócitos Mononucleares/microbiologia , Microvasos/microbiologia , Animais , Barreira Hematoencefálica/citologia , Brucella abortus/fisiologia , Brucelose/microbiologia , Retículo Endoplasmático/microbiologia , Endossomos/microbiologia , Células Endoteliais/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Cultura Primária de Células , Transcitose/fisiologia , Vacúolos/microbiologia
5.
PLoS Pathog ; 14(4): e1006981, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29630665

RESUMO

Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Endotélio Vascular/patologia , Proteínas de Membrana/metabolismo , Infecções Meningocócicas/complicações , Microvasos/patologia , Proteína C/metabolismo , Púrpura Fulminante/etiologia , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Aderência Bacteriana , Coagulação Sanguínea/fisiologia , Células Cultivadas , Receptor de Proteína C Endotelial/genética , Endotélio Vascular/metabolismo , Endotélio Vascular/microbiologia , Humanos , Proteínas de Membrana/genética , Infecções Meningocócicas/microbiologia , Microvasos/metabolismo , Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Proteína C/genética , Púrpura Fulminante/metabolismo , Púrpura Fulminante/patologia
6.
Virulence ; 8(8): 1808-1819, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29099305

RESUMO

Neisseria meningitidis is the causative agent of cerebrospinal meningitis and that of a rapidly progressing fatal septic shock known as purpura fulminans. Meningococcemia is characterized by bacterial adhesion to human endothelial cells of the microvessels. Host specificity has hampered studies on the role of blood vessels colonization in N. meningitidis associated pathogenesis. In this work, using a humanized model of SCID mice allowing the study of bacterial adhesion to human cells in an in vivo context we demonstrate that meningococcal colonization of human blood vessels is a prerequisite to the establishment of sepsis and lethality. To identify the molecular pathways involved in bacterial virulence, we performed transposon insertion site sequencing (Tn-seq) in vivo. Our results demonstrate that 36% of the genes that are important for growth in the blood of mice are dispensable when bacteria colonize human blood vessels, suggesting that human endothelial cells lining the blood vessels are feeding niches for N. meningitidis in vivo. Altogether, our work proposes a new paradigm for meningococcal virulence in which colonization of blood vessels is associated with metabolic adaptation and sustained bacteremia responsible for sepsis and subsequent lethality.


Assuntos
Bacteriemia/microbiologia , Infecções Meningocócicas/sangue , Infecções Meningocócicas/microbiologia , Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Animais , Bacteriemia/sangue , Aderência Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neisseria meningitidis/genética
7.
Traffic ; 18(11): 733-746, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28799243

RESUMO

Eukaryotic cells utilize multiple endocytic pathways for specific uptake of ligands or molecules, and these pathways are commonly hijacked by pathogens to enable host cell invasion. Escherichia coli K1, a pathogenic bacterium that causes neonatal meningitis, invades the endothelium of the blood-brain barrier, but the entry route remains unclear. Here, we demonstrate that the bacteria trigger an actin-mediated uptake route, stimulating fluid phase uptake, membrane ruffling and macropinocytosis. The route of uptake requires intact lipid rafts as shown by cholesterol depletion. Using a variety of perturbants we demonstrate that small Rho GTPases and their downstream effectors have a significant effect on bacterial invasion. Furthermore, clathrin-mediated endocytosis appears to play an indirect role in E. coli K1 uptake. The data suggest that the bacteria effect a complex interplay between the Rho GTPases to increase their chances of uptake by macropinocytosis into human brain microvascular endothelial cells.


Assuntos
Encéfalo/microbiologia , Células Endoteliais/microbiologia , Escherichia coli/patogenicidade , Microvasos/microbiologia , Pinocitose/fisiologia , Actinas/metabolismo , Translocação Bacteriana , Encéfalo/irrigação sanguínea , Linhagem Celular , Colesterol/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Endotélio Vascular/microbiologia , Escherichia coli/fisiologia , Humanos , Microvasos/metabolismo , Virulência
8.
J Clin Invest ; 127(6): 2249-2261, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28463232

RESUMO

Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.


Assuntos
Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Biespecíficos , Antígenos de Bactérias/imunologia , Carga Bacteriana , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Proteínas do Sistema Complemento/metabolismo , Avaliação Pré-Clínica de Medicamentos , Feminino , Células de Kupffer/microbiologia , Pulmão/irrigação sanguínea , Pulmão/microbiologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Microvasos/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fagocitose , Proteínas Citotóxicas Formadoras de Poros/imunologia , Infecções por Pseudomonas/imunologia , Receptores Fc/metabolismo
9.
Vet Pathol ; 54(4): 629-639, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28178428

RESUMO

Histophilus somni is a pathogenic gram-negative bacterium responsible for pneumonia and septicemia in cattle. Sequelae include infectious thrombotic meningoencephalitis (ITME), myocarditis, arthritis, and abortion. These syndromes are associated with widespread vasculitis and thrombosis, implicating a role for endothelium in pathogenesis. Histopathologic and immunohistochemical investigation of 10 natural cases of bovine H. somni myocarditis and 1 case of ITME revealed intravascular H. somni in large biofilm-like aggregates adherent to the luminal surface of microvascular endothelium. Ultrastructurally, bacterial communities were extracellular and closely associated with degenerating or contracted endothelial cells. Histophilus somni was identified by bacterial culture and/or immunohistochemistry. Western blots of the bacterial isolates revealed that they expressed the immunodominant protective 40 kDa OMP and immunoglobulin-binding protein A (IbpA) antigens. The latter is a large surface antigen and shed fibrillar antigen with multiple domains. The cytotoxic DR2Fic domain of IbpA was conserved as demonstrated by polymerase chain reaction. Treatment of endothelial cells in vitro with IbpA in crude culture supernatants or purified recombinant GST-IbpA DR2Fic (rDR2) cytotoxin induced retraction of cultured bovine brain microvascular endothelial cells. By contrast, no retraction of bovine endothelium was induced by mutant rDR2H/A with an inactive Fic motif or by a GST control, indicating that the cytotoxic DR2Fic motif plays an important role in endothelial cell retraction in vasculitis. The formation of biofilm-like aggregates by H. somni on bovine microvascular endothelium may be fundamental to its pathogenesis in heart and brain.


Assuntos
Encéfalo/patologia , Doenças dos Bovinos/microbiologia , Endotélio Vascular/patologia , Microvasos/patologia , Miocárdio/patologia , Infecções por Pasteurellaceae/veterinária , Pasteurellaceae , Animais , Western Blotting/veterinária , Encéfalo/microbiologia , Bovinos , Doenças dos Bovinos/patologia , Endotélio Vascular/microbiologia , Coração/microbiologia , Pulmão/microbiologia , Pulmão/patologia , Masculino , Microvasos/microbiologia , Infecções por Pasteurellaceae/patologia , Reação em Cadeia da Polimerase/veterinária
10.
Infect Immun ; 84(10): 3017-23, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481255

RESUMO

Meningococcal septic shock is associated with profound vasoplegia, early and severe myocardial dysfunction, and extended skin necrosis responsible for a specific clinical entity designated purpura fulminans (PF). PF represents 90% of fatal meningococcal infections. One characteristic of meningococcal PF is the myocardial dysfunction that occurs in the early phase of sepsis. Furthermore, hemodynamic studies have shown that the prognosis of meningococcal sepsis is directly related to the degree of impairment of cardiac contractility during the initial phase of the disease. To gain insight into a potential interaction of Neisseria meningitidis with the myocardial microvasculature, we modified a previously described humanized mouse model by grafting human myocardial tissue to SCID mice. We then infected the grafted mice with N. meningitides Using the humanized SCID mouse model, we demonstrated that N. meningitidis targets the human myocardial tissue vasculature, leading to the formation of blood thrombi, infectious vasculitis, and vascular leakage. These results suggest a novel mechanism of myocardial injury in the course of severe N. meningitidis sepsis that is likely to participate in primary myocardial dysfunction.


Assuntos
Coração/microbiologia , Infecções Meningocócicas/microbiologia , Microvasos/microbiologia , Animais , Bacteriemia/microbiologia , Modelos Animais de Doenças , Células Endoteliais/microbiologia , Células Endoteliais/patologia , Feminino , Humanos , Infecções Meningocócicas/patologia , Camundongos SCID , Miocárdio , Neisseria meningitidis , Choque Séptico/sangue , Vasculite/patologia , Trombose Venosa/patologia
11.
Oncotarget ; 6(32): 32456-67, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26440149

RESUMO

Invasion and traversal of the blood-brain barrier (BBB) by Mycobacterium tuberculosis cause meningeal tuberculosis (TB) in the central nervous system (CNS). Meningeal TB is a serious, often fatal disease that disproportionately affects young children. The mechanisms involved in CNS invasion by M. tuberculosis bacilli are poorly understood. In this study, we microscopically examined endosomal trafficking and measured survival of M. tuberculosis and M. bovis Bacille Calmette-Guérin (BCG) bacilli in murine brain microvascular endothelial cells (BMECs). The results show that both species internalize but do not replicate in BMECs in the absence of a cytotoxic response. Confocal microscopy indicates that bacilli-containing vacuoles are associated with the early endosomal marker, Rab5, late endosomal marker, Rab7, and lysosomal marker, LAMP2, suggesting that bacilli-containing endosomes mature into endolysosomes in BMECs. Our data also show that a subset of intracellular M. tuberculosis, but not BCG bacilli, escape into the cytoplasm to avoid rapid lysosomal killing. However, the intracellular mycobacteria examined cannot spread cell-to-cell in BMECs. Taken together, these data show that with the exception of the small terminal cytoplasmic population of bacilli, M. tuberculosis does not modulate intracellular trafficking in BMECs as occurs in macrophages and lung epithelial and endothelial cells.


Assuntos
Encéfalo/irrigação sanguínea , Permeabilidade Capilar , Células Endoteliais/microbiologia , Lisossomos/microbiologia , Microvasos/microbiologia , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium tuberculosis/crescimento & desenvolvimento , Animais , Linhagem Celular , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Camundongos , Viabilidade Microbiana , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microvasos/metabolismo , Microvasos/ultraestrutura , Mycobacterium bovis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Fatores de Tempo , Virulência , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
12.
J Thromb Haemost ; 13(10): 1743-56, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26190521

RESUMO

Sepsis, also known as septicemia, is one of the 10 leading causes of death worldwide. The rising tide of sepsis due to bacterial, fungal and viral infections cannot be stemmed by current antimicrobial therapies and supportive measures. New paradigms for the mechanism and resolution of sepsis and consequences for sepsis survivors are emerging. Consistent with Benjamin Franklin's dictum 'an ounce of prevention is worth a pound of cure', sepsis can be prevented by vaccinations against pneumococci and meningococci. Recently, the NIH NHLBI Panel redefined sepsis as 'severe endothelial dysfunction syndrome in response to intravascular and extravascular infections causing reversible or irreversible injury to the microcirculation responsible for multiple organ failure'. Microvascular endothelial injury underlies sepsis-associated hypotension, edema, disseminated intravascular coagulation, acute respiratory distress syndrome and acute kidney injury. Microbial genome products trigger 'genome wars' in sepsis that reprogram the human genome and culminate in a 'genomic storm' in blood and vascular cells. Sepsis can be averted experimentally by endothelial cytoprotection through targeting nuclear signaling that mediates inflammation and deranged metabolism. Endothelial 'rheostats' (e.g. inhibitors of NF-κB, A20 protein, CRADD/RAIDD protein and microRNAs) regulate endothelial signaling. Physiologic 'extinguishers' (e.g. suppressor of cytokine signaling 3) can be replenished through intracellular protein therapy. Lipid mediators (e.g. resolvin D1) hasten sepsis resolution. As sepsis cases rose from 387 330 in 1996 to 1.1 million in 2011, and are estimated to reach 2 million by 2020 in the US, mortality due to sepsis approaches that of heart attacks and exceeds deaths from stroke. More preventive vaccines and therapeutic measures are urgently needed.


Assuntos
Microcirculação , Microvasos/fisiopatologia , Sepse/prevenção & controle , Sepse/terapia , Animais , Predisposição Genética para Doença , Genoma Bacteriano , Genoma Fúngico , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Microvasos/metabolismo , Microvasos/microbiologia , Microvasos/virologia , Fenótipo , Fatores de Risco , Sepse/diagnóstico , Sepse/genética , Sepse/metabolismo , Sepse/mortalidade , Sepse/fisiopatologia , Transdução de Sinais , Resultado do Tratamento
13.
Am J Respir Cell Mol Biol ; 53(4): 459-70, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25693001

RESUMO

A major cause of death after influenza virus infection is lung injury due to a bacterial superinfection, yet the mechanism is unknown. Death has been attributed to virus-induced immunosuppression and bacterial overgrowth, but this hypothesis is based on data from the preantibiotic era and animal models that omit antimicrobial therapy. Because of diagnostic uncertainty, most patients with influenza receive antibiotics, making bacterial overgrowth unlikely. Respiratory failure after superinfection presents as acute respiratory distress syndrome, a disorder characterized by lung microvascular leak and edema. The objective of this study was to determine whether the influenza virus sensitizes the lung endothelium to leak upon exposure to circulating bacterial-derived molecular patterns from Staphylococcus aureus. In vitro as well as in vivo models of influenza followed by S. aureus superinfection were used. Molecular mechanisms were explored using molecular biology, knockout mice, and human autopsy specimens. Influenza virus infection sensitized human lung endothelium to leak when challenged with S. aureus, even at low doses of influenza and even when the pathogens were given days apart. Influenza virus increased endothelial expression of TNFR1 both in vitro and in intact lungs, a finding corroborated by human autopsy specimens of patients with influenza. Leak was recapitulated with protein A, a TNFR1 ligand, and sequential infection caused protein A-dependent loss of IκB, cleavage of caspases 8 and 3, and lung endothelial apoptosis. Mice infected sequentially with influenza virus and S. aureus developed significantly increased lung edema that was protein A and TNFR1 dependent. Influenza virus primes the lung endothelium to leak, predisposing patients to acute respiratory distress syndrome upon exposure to S. aureus.


Assuntos
Endotélio Vascular/metabolismo , Influenza Humana/metabolismo , Microvasos/metabolismo , Infecções Estafilocócicas/metabolismo , Animais , Apoptose , Permeabilidade Capilar , Células Cultivadas , Endotélio Vascular/microbiologia , Endotélio Vascular/patologia , Humanos , Vírus da Influenza A Subtipo H3N2/fisiologia , Influenza Humana/patologia , Influenza Humana/virologia , Pulmão/irrigação sanguínea , Camundongos , Camundongos Knockout , Microvasos/microbiologia , Microvasos/patologia , NF-kappa B/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Infecções Estafilocócicas/patologia , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/fisiologia , Regulação para Cima
14.
Microb Pathog ; 78: 29-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25450888

RESUMO

Jorge Lobo's disease is a rare mycosis characterized by chronic inflammation, which causes skin lesions in the absence of visceral dissemination. The disease occurs mainly in hot and humid climates and most cases have been registered in the Brazilian Amazon region. This study investigated possible microvascular alterations in skin lesions caused by infection with Lacazia loboi which may interfere with the clinical progression of the disease. Immunohistochemistry was used to evaluate the density of blood and lymphatic vessels, as well as expression of the cell adhesion molecules ICAM-1, VCAM-1 and E-selectin. The results showed a reduced number of blood (62.66 ± 20.30 vessels/mm(2)) and lymphatic vessels (3.55 ± 5.84 vessels/mm(2)) in Jorge Lobo's disease when compared to control skin (169.66 ± 66.38 blood vessels/mm(2) and 8 ± 2.17 lymphatic vessels/mm(2)). There were a larger number of vessels expressing ICAM-1 (27.58 ± 15.32 vessels/mm(2)) and VCAM-1 (7.55 ± 6.2 vessels/mm(2)). No difference was observed in the expression of E-selectin (4.66 ± 11 vessels/mm(2)). Taken together, the results indicate changes in the local microvasculature which may interfere with the development of an efficient cell-mediated immune response and may explain restriction of the fungus to the site of injury.


Assuntos
Células Endoteliais/patologia , Lacazia/fisiologia , Lobomicose/patologia , Microvasos/patologia , Pele/irrigação sanguínea , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Brasil , Selectina E/genética , Selectina E/metabolismo , Células Endoteliais/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Lobomicose/genética , Lobomicose/metabolismo , Lobomicose/microbiologia , Masculino , Microvasos/metabolismo , Microvasos/microbiologia , Pessoa de Meia-Idade , Pele/metabolismo , Pele/patologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Adulto Jovem
15.
Caries Res ; 48(6): 534-48, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24993646

RESUMO

This study investigated the distribution patterns of glial networks disclosed by reactivity for glial fibrillary acidic protein (GFAP) and S100B in healthy and carious human teeth. The objective was to determine the assembly and collapse of glial networks in response to encroaching infection. 15 healthy and 37 carious posterior teeth from adults were studied. Immediately after extraction, teeth were cleaned and vertically split and the half with pulp fixed and prepared for resin or frozen sections. Sections were stained with toluidine blue and for immunofluorescence, with observation by confocal laser microscopy and analysis by ImageJ software. Carious teeth were subdivided into three groups according to degree of carious involvement: microbial penetration through enamel (stage A), extension into dentin (stage B) and advanced penetration into dentin but without invasion of underlying pulp tissue (stage C). In stage A lesions there was marked increase in glial networks in dental pulp tissue that extended beyond the zone of microbial invasion. This response was maintained in stage B lesions. In advanced stage C lesions these networks were degraded in the zone of invasion in association with failure to contain infection. Cells expressing the glial markers GFAP and S100B showed a response to initial microbial invasion of dentin by increase in number and altered anatomical arrangement. The late stage of dentinal caries was marked by collapse of these networks in the region adjacent to advancing bacteria. This behaviour is important for understanding and explaining the defensive response of the neurosensory peripheral dental pulp apparatus to infection.


Assuntos
Coinfecção/microbiologia , Cárie Dentária/microbiologia , Dentina/inervação , Neuroglia/patologia , Adulto , Astrócitos/microbiologia , Astrócitos/patologia , Biomarcadores/análise , Colágeno Tipo IV/análise , Corantes , Esmalte Dentário/microbiologia , Polpa Dentária/irrigação sanguínea , Polpa Dentária/inervação , Dentina/microbiologia , Progressão da Doença , Secções Congeladas , Proteína Glial Fibrilar Ácida/análise , Humanos , Microvasos/microbiologia , Microvasos/patologia , Pessoa de Meia-Idade , Odontoblastos/microbiologia , Odontoblastos/patologia , Inclusão em Plástico , Subunidade beta da Proteína Ligante de Cálcio S100/análise , Células Receptoras Sensoriais/microbiologia , Cloreto de Tolônio , Vimentina/análise , Adulto Jovem
16.
PLoS Pathog ; 10(6): e1004160, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945304

RESUMO

The interaction with brain endothelial cells is central to the pathogenicity of Neisseria meningitidis infections. Here, we show that N. meningitidis causes transient activation of acid sphingomyelinase (ASM) followed by ceramide release in brain endothelial cells. In response to N. meningitidis infection, ASM and ceramide are displayed at the outer leaflet of the cell membrane and condense into large membrane platforms which also concentrate the ErbB2 receptor. The outer membrane protein Opc and phosphatidylcholine-specific phospholipase C that is activated upon binding of the pathogen to heparan sulfate proteoglycans, are required for N. meningitidis-mediated ASM activation. Pharmacologic or genetic ablation of ASM abrogated meningococcal internalization without affecting bacterial adherence. In accordance, the restricted invasiveness of a defined set of pathogenic isolates of the ST-11/ST-8 clonal complex into brain endothelial cells directly correlated with their restricted ability to induce ASM and ceramide release. In conclusion, ASM activation and ceramide release are essential for internalization of Opc-expressing meningococci into brain endothelial cells, and this segregates with invasiveness of N. meningitidis strains.


Assuntos
Encéfalo/irrigação sanguínea , Ceramidas/metabolismo , Endotélio Vascular/microbiologia , Interações Hospedeiro-Patógeno , Neisseria meningitidis/patogenicidade , Esfingomielina Fosfodiesterase/metabolismo , Regulação para Cima , Aderência Bacteriana/efeitos dos fármacos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/microbiologia , Linhagem Celular Transformada , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/enzimologia , Microdomínios da Membrana/metabolismo , Meningite Meningocócica/enzimologia , Meningite Meningocócica/metabolismo , Meningite Meningocócica/microbiologia , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/microbiologia , Mutação , Neisseria meningitidis/fisiologia , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Propriedades de Superfície/efeitos dos fármacos , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo , Regulação para Cima/efeitos dos fármacos
17.
Thromb Res ; 133 Suppl 1: S35-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24759139

RESUMO

Microvascular thrombosis indicates a pathological occlusion of microvessels by fibrin- and/or platelet-rich thrombi. It is observed during systemic infections, cancer, myocardial infarction, stroke, neurodegenerative diseases and in thrombotic microangiopathies. Microvessel thrombosis can cause greatly differing symptoms that range from limited changes in plasma coagulation markers to severe multi-organ failure. Because microvessel thrombi are difficult to detect and often occur only transiently, their importance for disease development and host biology is likely markedly under-appreciated. Recently, clear indications for a biological basis of microvascular thrombosis have been obtained. During systemic infections microvessel thrombosis can mediate an intravascular innate immune response (immunothrombosis). This biological form of thrombosis is based on the generation of fibrin inside blood vessels and is critically triggered by neutrophils and their interactions with platelets which result in the release of neutrophil extracellular traps (extracellular nucleosomes). Immunothrombosis is critically supported by neutrophil elastase and the activator molecules of blood coagulation tissue factor and factor XII. Identification of the biological driving forces of microvascular thrombosis should help to elucidate the mechanisms promoting pathological vessel occlusions in both microvessels and large vessels.


Assuntos
Microvasos/imunologia , Microvasos/patologia , Trombose/imunologia , Trombose/patologia , Animais , Coagulação Sanguínea , Fibrina/metabolismo , Humanos , Imunidade Inata , Microvasos/microbiologia , Trombose/sangue , Trombose/microbiologia
18.
J Vis Exp ; (86)2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24747976

RESUMO

Neisseria meningitidis causes a severe, frequently fatal sepsis when it enters the human blood stream. Infection leads to extensive damage of the blood vessels resulting in vascular leak, the development of purpuric rashes and eventual tissue necrosis. Studying the pathogenesis of this infection was previously limited by the human specificity of the bacteria, which makes in vivo models difficult. In this protocol, we describe a humanized model for this infection in which human skin, containing dermal microvessels, is grafted onto immunocompromised mice. These vessels anastomose with the mouse circulation while maintaining their human characteristics. Once introduced into this model, N. meningitidis adhere exclusively to the human vessels, resulting in extensive vascular damage, inflammation and in some cases the development of purpuric rash. This protocol describes the grafting, infection and evaluation steps of this model in the context of N. meningitidis infection. The technique may be applied to numerous human specific pathogens that infect the blood stream.


Assuntos
Modelos Animais de Doenças , Infecções Meningocócicas/microbiologia , Microvasos/transplante , Neisseria meningitidis/patogenicidade , Transplante de Pele/métodos , Pele/irrigação sanguínea , Transplante Heterólogo/métodos , Animais , Xenoenxertos , Humanos , Camundongos , Microvasos/microbiologia
19.
J Infect Dis ; 208(10): 1590-7, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23840047

RESUMO

Neisseria meningitidis is a strict human pathogen that closely interacts with human endothelial cells via type IV pili in vitro. To decipher whether this interaction plays a role in vivo, we set up an experimental model of fulminant meningococcemia in human skin grafted SCID mice using the wild-type strain 2C4.3. Human skin and mouse tissues were sampled 24 hours after bacterial challenge for histopathology, immunohistochemistry and ultrastructural analysis. In all infected mice, N. meningitidis targeted the human vasculature, leading to bacterial and blood thrombi, infectious vasculitis and vascular leakage. Mouse vessels, including brain vessels, remained unaffected by the infectious and thrombotic process, and a nonpiliated Δ pilE derivative of 2C4.3 failed to target human graft vessels and to induce vascular damages. These data demonstrate that N. meningitidis targets human endothelial cells in vivo and that this interaction triggers the vascular damages that characterize purpura fulminans.


Assuntos
Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Púrpura Fulminante/etiologia , Púrpura Fulminante/patologia , Animais , Aderência Bacteriana , Células Endoteliais/microbiologia , Células Endoteliais/patologia , Endotélio Vascular/microbiologia , Endotélio Vascular/patologia , Feminino , Fímbrias Bacterianas/fisiologia , Xenoenxertos , Humanos , Infecções Meningocócicas/complicações , Infecções Meningocócicas/microbiologia , Camundongos , Microvasos/patologia , Pele/irrigação sanguínea , Pele/patologia , Transplante de Pele
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA