Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 622(7983): 611-618, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37699522

RESUMO

Clostridioides difficile infection (CDI) is a major cause of healthcare-associated gastrointestinal infections1,2. The exaggerated colonic inflammation caused by C. difficile toxins such as toxin B (TcdB) damages tissues and promotes C. difficile colonization3-6, but how TcdB causes inflammation is unclear. Here we report that TcdB induces neurogenic inflammation by targeting gut-innervating afferent neurons and pericytes through receptors, including the Frizzled receptors (FZD1, FZD2 and FZD7) in neurons and chondroitin sulfate proteoglycan 4 (CSPG4) in pericytes. TcdB stimulates the secretion of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) from neurons and pro-inflammatory cytokines from pericytes. Targeted delivery of the TcdB enzymatic domain, through fusion with a detoxified diphtheria toxin, into peptidergic sensory neurons that express exogeneous diphtheria toxin receptor (an approach we term toxogenetics) is sufficient to induce neurogenic inflammation and recapitulates major colonic histopathology associated with CDI. Conversely, mice lacking SP, CGRP or the SP receptor (neurokinin 1 receptor) show reduced pathology in both models of caecal TcdB injection and CDI. Blocking SP or CGRP signalling reduces tissue damage and C. difficile burden in mice infected with a standard C. difficile strain or with hypervirulent strains expressing the TcdB2 variant. Thus, targeting neurogenic inflammation provides a host-oriented therapeutic approach for treating CDI.


Assuntos
Toxinas Bacterianas , Clostridioides difficile , Inflamação Neurogênica , Neurônios Aferentes , Pericitos , Animais , Camundongos , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/farmacologia , Peptídeo Relacionado com Gene de Calcitonina/antagonistas & inibidores , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Inflamação Neurogênica/induzido quimicamente , Inflamação Neurogênica/microbiologia , Inflamação Neurogênica/patologia , Pericitos/efeitos dos fármacos , Pericitos/microbiologia , Pericitos/patologia , Receptores da Neurocinina-1/metabolismo , Substância P/antagonistas & inibidores , Substância P/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/microbiologia , Neurônios Aferentes/patologia , Mediadores da Inflamação/metabolismo , Ceco/efeitos dos fármacos , Ceco/metabolismo , Transdução de Sinais/efeitos dos fármacos
2.
Infect Immun ; 82(2): 851-63, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478098

RESUMO

Klebsiella pneumoniae has become an important pathogen in recent years. Although most cases of K. pneumoniae endogenous endophthalmitis occur via hematogenous spread, it is not yet clear which microbial and host factors are responsible for the ability of K. pneumoniae to cross the blood-retinal barrier (BRB). In the present study, we show that in an in vitro model of BRB based on coculturing primary bovine retinal endothelial cells (BREC) and primary bovine retinal pericytes (BRPC), K. pneumoniae infection determines changes of transendothelial electrical resistance (TEER) and permeability to sodium fluorescein. In the coculture model, bacteria are able to stimulate the enzyme activities of endothelial cytosolic and Ca(2+)-independent phospholipase A2s (cPLA2 and iPLA2). These results were confirmed by the incremental expression of cPLA2, iPLA2, cyclo-oxygenase-1 (COX1), and COX2 in BREC, as well as by cPLA2 phosphorylation. In supernatants of K. pneumoniae-stimulated cocultures, increases in prostaglandin E2 (PGE2), interleukin-6 (IL-6), IL-8, and vascular endothelial growth factor (VEGF) production were found. Incubation with K. pneumoniae in the presence of arachidonoyl trifluoromethyl ketone (AACOCF3) or bromoenol lactone (BEL) caused decreased PGE2 and VEGF release. Scanning electron microscopy and transmission electron microscopy images of BREC and BRPC showed adhesion of K. pneumoniae to the cells, but no invasion occurred. K. pneumoniae infection also produced reductions in pericyte numbers; transfection of BREC cocultured with BRPC and of human retinal endothelial cells (HREC) cocultured with human retinal pericytes (HRPC) with small interfering RNAs (siRNAs) targeted to cPLA2 and iPLA2 restored the pericyte numbers and the TEER and permeability values. Our results show the proinflammatory effect of K. pneumoniae on BREC, suggest a possible mechanism by which BREC and BRPC react to the K. pneumoniae infection, and may provide physicians and patients with new ways of fighting blinding diseases.


Assuntos
Barreira Hematorretiniana/microbiologia , Barreira Hematorretiniana/patologia , Células Endoteliais/microbiologia , Inflamação/microbiologia , Inflamação/fisiopatologia , Klebsiella pneumoniae/imunologia , Pericitos/microbiologia , Animais , Bovinos , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/fisiologia , Perfilação da Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Klebsiella pneumoniae/patogenicidade , Pericitos/fisiologia , Permeabilidade
3.
Cell Microbiol ; 15(8): 1367-84, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23421875

RESUMO

The key aspect of neonatal meningitis is related to the ability of pathogens to invade the blood-brain barrier (BBB) and to penetrate the central nervous system. In the present study we show that, in an in vitro model of BBB, on the basis of co-culturing primary bovine brain endothelial cells (BBEC) and primary bovine retinal pericytes (BRPC), Escherichia coli infection determines changes of transendothelial electrical resistance (TEER) and permeability (Pe) to sodium fluorescein. In the co-culture model, within BBEC, bacteria are able to stimulate cytosolic and Ca(2+)-independent phospholipase A2 (cPLA2 and iPLA2 ) enzyme activities. In supernatants of E. coli-stimulated co-cultures, an increase in prostaglandins (PGE2) and VEGF production in comparison with untreated co-cultures were found. Incubation with E. coli in presence of AACOCF3 or BEL caused a decrease of PGE2 and VEGF release. SEM and TEM images of BBEC and BRPC showed E. coli adhesion to BBEC and BRPC but only in BBEC the invasion occurs. VEGFR-1 but not VEGFR-2 blockade by the specific antibody reduced E. coli invasion in BBEC. In our model of BBB infection, a significant loss of BRPC was observed. Following VEGFR-1, but not VEGFR-2 blockade, or in presence of AACOCF3 or BEL, elevated TEER values, reduced permeability and BRPC loss were found. These data suggest that VEGFR-1 negatively regulates BRPC survival and its blockade protects the barrier integrity. PGs and VEGF could exert a biological effect on BBB, probably by BRPC coverage ablation, thus increasing BBB permeability. Our results show the role played by the BBEC as well as BRPC during a bacterial attack on BBB. A better understanding of the mechanisms by which E. coli enter the nervous system and how bacteria alter the communication between endothelial cells and pericytes may provide exciting new insight for clinical intervention.


Assuntos
Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/fisiopatologia , Endotélio Vascular/patologia , Infecções por Escherichia coli/fisiopatologia , Escherichia coli/patogenicidade , Pericitos/patologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Aderência Bacteriana/fisiologia , Barreira Hematoencefálica/patologia , Bovinos , Permeabilidade da Membrana Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Dinoprostona/metabolismo , Modelos Animais de Doenças , Impedância Elétrica , Endotélio Vascular/microbiologia , Escherichia coli/fisiologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Fluoresceína , Corantes Fluorescentes , Pericitos/microbiologia , Fosfolipases A2/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/imunologia
4.
Med Microbiol Immunol ; 202(2): 143-51, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23184416

RESUMO

Angiogenesis is an important physiological and pathological process. Bartonella is the only genus of bacteria known to induce pathological angiogenesis in the mammalian host. Bartonella-induced angiogenesis leads to the formation of vascular tumors including verruga peruana and bacillary angiomatosis. The mechanism of Bartonella-induced angiogenesis is not completely understood. Pericytes, along with endothelial cells, play an important role in physiological angiogenesis, and their role in tumor angiogenesis has been extensively studied. Abnormal signaling between endothelial cells and pericytes contributes to tumor angiogenesis and metastasis; however, the role of pericytes in Bartonella-induced angiogenesis is not known. In this study, after infecting human brain vascular pericytes (HBVPs) with Bartonella henselae, we found that these bacteria were able to invade HBVPs and that bacterial infection resulted in decreased pericyte proliferation and increased pericyte production of vascular endothelial growth factor (VEGF) when compared to the uninfected control cells. In the context of pathological angiogenesis, reduced pericyte coverage, accompanied by increased VEGF production, may promote endothelial cell proliferation and the formation of new vessels.


Assuntos
Angiomatose Bacilar/microbiologia , Bartonella henselae/fisiologia , Encéfalo/microbiologia , Pericitos/microbiologia , Angiomatose Bacilar/metabolismo , Apoptose , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células , Células Cultivadas , Humanos , Pericitos/metabolismo , Pericitos/ultraestrutura , Fator A de Crescimento do Endotélio Vascular/biossíntese
5.
Am J Pathol ; 178(6): 2897-909, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21550017

RESUMO

Blood vessel leakiness is an early, transient event in acute inflammation but can also persist as vessels undergo remodeling in sustained inflammation. Angiopoietin/Tie2 signaling can reduce the leakiness through changes in endothelial cells. The role of pericytes in this action has been unknown. We used the selective PDGF-B-blocking oligonucleotide aptamer AX102 to determine whether disruption of pericyte-endothelial crosstalk alters vascular leakiness or remodeling in the airways of mice under four different conditions: i) baseline, ii) acute inflammation induced by bradykinin, iii) sustained inflammation after 7-day infection by the respiratory pathogen Mycoplasma pulmonis, or iv) leakage after bradykinin challenge in the presence of vascular stabilization by the angiopoietin-1 (Ang1) mimic COMP-Ang1 for 7 days. AX102 reduced pericyte coverage but did not alter the leakage of microspheres from tracheal blood vessels at baseline or after bradykinin; however, AX102 exaggerated leakage at 7 days after M. pulmonis infection and increased vascular remodeling and disease severity at 14 days. AX102 also abolished the antileakage effect of COMP-Ang1 at 7 days. Together, these findings show that pericyte contributions to endothelial stability have greater dependence on PDGF-B during the development of sustained inflammation, when pericyte dynamics accompany vascular remodeling, than under baseline conditions or in acute inflammation. The findings also show that the antileakage action of Ang1 requires PDGF-dependent actions of pericytes in maintaining endothelial stability.


Assuntos
Angiopoietina-1/metabolismo , Inflamação/patologia , Pericitos/patologia , Traqueia/irrigação sanguínea , Traqueia/patologia , Actinas/metabolismo , Animais , Aptâmeros de Nucleotídeos/farmacologia , Bradicinina/farmacologia , Contagem de Células , Forma Celular/efeitos dos fármacos , Desmina/metabolismo , Inflamação/complicações , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Infecções por Mycoplasma/complicações , Infecções por Mycoplasma/patologia , Mycoplasma pulmonis/efeitos dos fármacos , Mycoplasma pulmonis/fisiologia , Pericitos/efeitos dos fármacos , Pericitos/microbiologia , Proteínas Proto-Oncogênicas c-sis/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-sis/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Traqueia/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...