Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
FASEB J ; 32(1): 26-36, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28855276

RESUMO

Leukocytes express formyl-peptide receptors (FPRs), which sense microbe-associated molecular pattern (MAMP) molecules, leading to leukocyte chemotaxis and activation. We recently demonstrated that phenol-soluble modulin (PSM) peptides from highly pathogenic Staphylococcus aureus are efficient ligands for the human FPR2. How PSM detection by FPR2 impacts on the course of S. aureus infections has remained unknown. We characterized the specificity of mouse FPR2 (mFpr2) using a receptor-transfected cell line, homeobox b8 (Hoxb8), and primary neutrophils isolated from wild-type (WT) or mFpr2-/- mice. The influx of leukocytes into the peritoneum of WT and mFpr2-/- mice was analyzed. We demonstrate that mFpr2 is specifically activated by PSMs in mice, and they represent the first secreted pathogen-derived ligands for the mFpr2. Intraperitoneal infection with S. aureus led to lower numbers of immigrated leukocytes in mFpr2-/- compared with WT mice at 3 h after infection, and this difference was not observed when mice were infected with an S. aureus PSM mutant. Our data support the hypothesis that the mFpr2 is the functional homolog of the human FPR2 and that a mouse infection model represents a suitable model for analyzing the role of PSMs during infection. PSM recognition by mFpr2 shapes leukocyte influx in local infections, the typical infections caused by S. aureus-Weiss, E., Hanzelmann, D., Fehlhaber, B., Klos, A., von Loewenich, F. D., Liese, J., Peschel, A., Kretschmer, D. Formyl-peptide receptor 2 governs leukocyte influx in local Staphylococcus aureus infections.


Assuntos
Leucócitos/imunologia , Receptores de Formil Peptídeo/imunologia , Receptores de Lipoxinas/imunologia , Infecções Estafilocócicas/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/imunologia , Sinalização do Cálcio/imunologia , Degranulação Celular/imunologia , Linhagem Celular , Movimento Celular/imunologia , Modelos Animais de Doenças , Feminino , Genes Bacterianos , Proteínas de Homeodomínio/imunologia , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Neutrófilos/imunologia , Receptores de Formil Peptídeo/deficiência , Receptores de Formil Peptídeo/genética , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia
2.
Kidney Int ; 94(4): 741-755, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29935951

RESUMO

Severe ischemia reperfusion injury (IRI) results in rapid complement activation, acute kidney injury and progressive renal fibrosis. Little is known about the roles of the C5aR1 and C5aR2 complement receptors in IRI. In this study C5aR1-/- and C5aR2-/- mice were compared to the wild type in a renal IRI model leading to renal fibrosis. C5a receptor expression, kidney morphology, inflammation, and fibrosis were measured in different mouse strains one, seven and 21 days after IRI. Renal perfusion was evaluated by functional magnetic resonance imaging. Protein abundance and phosphorylation were assessed with high content antibody microarrays and Western blotting. C5aR1 and C5aR2 were increased in damaged tubuli and even more in infiltrating leukocytes after IRI in kidneys of wild-type mice. C5aR1-/- and C5aR2-/- animals developed less IRI-induced inflammation and showed better renal perfusion than wild-type mice following IRI. C5aR2-/- mice, in particular, had enhanced tubular and capillary regeneration with less renal fibrosis. Anti-inflammatory IL-10 and the survival/growth kinase AKT levels were especially high in kidneys of C5aR2-/- mice following IRI. LPS caused bone marrow-derived macrophages from C5aR2-/- mice to release IL-10 and to express the stress response enzyme heme oxygenase-1. Thus, C5aR1 and C5aR2 have overlapping actions in which the kidneys of C5aR2-/- mice regenerate better than those in C5aR1-/- mice following IRI. This is mediated, at least in part, by differential production of IL-10, heme oxygenase-1 and AKT.


Assuntos
Heme Oxigenase-1/metabolismo , Interleucina-10/metabolismo , Túbulos Renais/patologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor da Anafilatoxina C5a/genética , Traumatismo por Reperfusão/genética , Animais , Proliferação de Células/genética , Células Cultivadas , Células Epiteliais , Fibrose , Inflamação/etiologia , Rim/diagnóstico por imagem , Túbulos Renais/metabolismo , Túbulos Renais/fisiopatologia , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Imageamento por Ressonância Magnética , Camundongos , Camundongos Knockout , Imagem de Perfusão , Fosforilação , Fatores de Proteção , Receptor da Anafilatoxina C5a/metabolismo , Regeneração/genética , Traumatismo por Reperfusão/complicações , Regulação para Cima
3.
Pharmacol Rev ; 65(1): 500-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23383423

RESUMO

The activation of the complement cascade, a cornerstone of the innate immune response, produces a number of small (74-77 amino acid) fragments, originally termed anaphylatoxins, that are potent chemoattractants and secretagogues that act on a wide variety of cell types. These fragments, C5a, C4a, and C3a, participate at all levels of the immune response and are also involved in other processes such as neural development and organ regeneration. Their primary function, however, is in inflammation, so they are important targets for the development of antiinflammatory therapies. Only three receptors for complement peptides have been found, but there are no satisfactory antagonists as yet, despite intensive investigation. In humans, there is a single receptor for C3a (C3a receptor), no known receptor for C4a, and two receptors for C5a (C5a1 receptor and C5a2 receptor). The most recently characterized receptor, the C5a2 receptor (previously known as C5L2 or GPR77), has been regarded as a passive binding protein, but signaling activities are now ascribed to it, so we propose that it be formally identified as a receptor and be given a name to reflect this. Here, we describe the complex biology of the complement peptides, introduce a new suggested nomenclature, and review our current knowledge of receptor pharmacology.


Assuntos
Receptores de Complemento/imunologia , Animais , Proteínas do Sistema Complemento/imunologia , Humanos , Peptídeos/imunologia
4.
J Infect Dis ; 209(8): 1269-78, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24273177

RESUMO

BACKGROUND: The complement system protects against extracellular pathogens and links innate and adaptive immunity. In this study, we investigated the anaphylatoxin C3a receptor (C3aR) in Chlamydia psittaci lung infection and elucidated C3a-dependent adaptive immune mechanisms. METHODS: Survival, body weight, and clinical score were monitored in primary mouse infection and after serum transfer. Bacterial load, histology, cellular distribution, cytokines, antibodies, and lymphocytes were analyzed. RESULTS: C3aR(-/-) mice showed prolonged pneumonia with decreased survival, lower weight, and higher clinical score. Compared to wild-type mice bacterial clearance was impaired, and inflammatory parameters were increased. In lung-draining lymph nodes of C3aR(-/-) mice the total number of B cells, CD4(+) T cells, and Chlamydia-specific IFN-γ(+) (CD4(+) or CD8(+)) cells was reduced upon infection, and the mice were incapable of Chlamydia-specific immunoglobulin M or immunoglobulin G production. Performed before infection, transfer of hyperimmune serum prolonged survival of C3aR(-/-) mice. CONCLUSIONS: C3a and its receptor are critical for defense against C. psittaci in mouse lung infection. In this model, C3a acts via its receptor as immune modulator. Enhancement of specific B and T cell responses upon infection with an intracellular bacterium were identified as hitherto unknown features of C3a/C3aR. These new functions might be of general immunological importance.


Assuntos
Imunidade Adaptativa/imunologia , Infecções por Chlamydophila/prevenção & controle , Chlamydophila psittaci/patogenicidade , Pulmão/microbiologia , Pneumonia Bacteriana/prevenção & controle , Receptores de Complemento/fisiologia , Linfócitos T/imunologia , Animais , Anticorpos Antibacterianos/sangue , Infecções por Chlamydophila/imunologia , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Masculino , Camundongos , Pneumonia Bacteriana/imunologia
5.
Infect Immun ; 82(8): 3154-63, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24842924

RESUMO

Hydrosalpinx is a pathological hallmark of tubal infertility associated with chlamydial infection. However, the mechanisms of hydrosalpinx remain unknown. Here, we report that complement factor 5 (C5) contributes significantly to chlamydial induction of hydrosalpinx. Mice lacking C5 (C5(-/-)) failed to develop any hydrosalpinx, while ∼42% of the corresponding wild-type mice (C5(+/+)) did so following intravaginal infection with Chlamydia muridarum. Surprisingly, deficiency in C3 (C3(-/-)), an upstream component of the complement system, did not affect mouse susceptibility to chlamydial induction of hydrosalpinx. Interestingly, C5 activation was induced by chlamydial infection in oviducts of C3(-/-) mice, explaining why the C3(-/-) mice remained susceptible to chlamydial induction of hydrosalpinx. Similar levels of live chlamydial organisms were recovered from oviduct tissues of both C5(-/-) and C5(+/+) mice, suggesting that C5 deficiency did not affect C. muridarum ascending infection. Furthermore, C5(-/-) mice were still more resistant to hydrosalpinx induction than C5(+/+) mice, even when live C. muridarum organisms were directly delivered into the upper genital tract, both confirming the role of C5 in promoting hydrosalpinx and indicating that the C5-facilitated hydrosalpinx was not due to enhancement of ascending infection. The C5(-/-) mice displayed significantly reduced lumenal inflammatory infiltration and cytokine production in oviduct tissue, suggesting that C5 may contribute to chlamydial induction of hydrosalpinx by enhancing inflammatory responses.


Assuntos
Infecções por Chlamydia/microbiologia , Infecções por Chlamydia/patologia , Chlamydia muridarum/imunologia , Complemento C3/imunologia , Complemento C5/imunologia , Salpingite/microbiologia , Salpingite/patologia , Animais , Complemento C5/genética , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Knockout
6.
Infect Immun ; 82(6): 2460-71, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24686060

RESUMO

Virulent Streptococcus suis serotype 2 strains are invasive extracellular bacteria causing septicemia and meningitis in piglets and humans. One objective of this study was to elucidate the function of complement in innate immune defense against S. suis. Experimental infection of wild-type (WT) and C3(-/-) mice demonstrated for the first time that the complement system protects naive mice against invasive mucosal S. suis infection. S. suis WT but not an unencapsulated mutant caused mortality associated with meningitis and other pathologies in C3(-/-) mice. The capsule contributed also substantially to colonization of the upper respiratory tract. Experimental infection of C3(-/-) mice with a suilysin mutant indicated that suilysin expression facilitated an early disease onset and the pathogenesis of meningitis. Flow cytometric analysis revealed C3 antigen deposition on the surface of ca. 40% of S. suis WT bacteria after opsonization with naive WT mouse serum, although to a significantly lower intensity than on the unencapsulated mutant. Ex vivo multiplication in murine WT and C3(-/-) blood depended on capsule but not suilysin expression. Interestingly, S. suis invasion of inner organs was also detectable in C5aR(-/-) mice, suggesting that chemotaxis and activation of immune cells via the anaphylatoxin receptor C5aR is, in addition to opsonization, a further important function of the complement system in defense against mucosal S. suis infection. In conclusion, we unequivocally demonstrate here the importance of complement against mucosal S. suis serotype 2 infection and that the capsule of this pathogen is also involved in escape from complement-independent immunity.


Assuntos
Cápsulas Bacterianas/fisiologia , Proteínas do Sistema Complemento/fisiologia , Proteínas Hemolisinas/fisiologia , Streptococcus suis/fisiologia , Anafilatoxinas/fisiologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Interações Hospedeiro-Patógeno/fisiologia , Imunidade Inata/fisiologia , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cavidade Nasal/microbiologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/mortalidade , Streptococcus suis/patogenicidade , Virulência
7.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 6): 1704-17, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24914981

RESUMO

Complement is an ancient part of the innate immune system that plays a pivotal role in protection against invading pathogens and helps to clear apoptotic and necrotic cells. Upon complement activation, a cascade of proteolytic events generates the complement effectors, including the anaphylatoxins C3a and C5a. Signalling through their cognate G-protein coupled receptors, C3aR and C5aR, leads to a wide range of biological events promoting inflammation at the site of complement activation. The function of anaphylatoxins is regulated by circulating carboxypeptidases that remove their C-terminal arginine residue, yielding C3a-desArg and C5a-desArg. Whereas human C3a and C3a-desArg adopt a canonical four-helix bundle fold, the conformation of human C5a-desArg has recently been described as a three-helix bundle. Here, the crystal structures of an antagonist version of human C5a, A8(Δ71-73), and of murine C5a and C5a-desArg are reported. Whereas A8(Δ71-73) adopts a three-helix bundle conformation similar to human C5a-desArg, the two murine proteins form a four-helix bundle. A cell-based functional assay reveals that murine C5a-desArg, in contrast to its human counterpart, exerts the same level of activition as murine C5a on its cognate receptor. The role of the different C5a conformations is discussed in relation to the differential activation of C5a receptors across species.


Assuntos
Anafilatoxinas/química , Anafilatoxinas/fisiologia , Complemento C5a/metabolismo , Animais , Cristalização , Humanos , Espectrometria de Massas , Camundongos , Modelos Moleculares
8.
Immunol Cell Biol ; 92(7): 631-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24777312

RESUMO

The complement system is a major component of our innate immune system, in which the complement proteins C5a and C5a-des Arg bind to two G-protein-coupled receptors: namely, the C5a receptor (C5a1) and C5a receptor like-2 receptor (C5a2, formerly called C5L2). Recently, it has been demonstrated that C5a, but not C5a-des Arg, upregulates heteromer formation between C5a1 and C5a2, leading to an increase in IL-10 release from human monocyte-derived macrophages (HMDMs). A bioluminescence resonance energy transfer (BRET) assay was used to assess the recruitment of ß-arrestins by C5a and C5a-des Arg at the C5a1 and C5a2 receptors. C5a demonstrated elevated ß-arrestin 2 recruitment levels in comparison with C5a-des Arg, whereas no significant difference was observed at C5a2. A constitutive complex that formed between ß-arrestin 2 and C5a2 accounted for half of the BRET signal observed. Interestingly, both C5a and C5a-des Arg exhibited higher potency for ß-arrestin 2 recruitment via C5a2, indicating preference for C5a2 over C5a1. When C5a was tested in a functional ERK1/2 assay in HMDMs, inhibition of ERK1/2 was observed only at concentrations at or above the EC50 for heteromer formation. This suggested that increased recruitment of the ß-arrestin-C5a2 complex at these C5a concentrations might have an inhibitory role on C5a1 signaling through ERK1/2. An improved understanding of C5a2 modulation of signaling in acute inflammation could be of benefit in the development of ligands for conditions such as sepsis.


Assuntos
Arrestinas/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , Receptores de Quimiocinas/metabolismo , Arrestinas/genética , Linhagem Celular , Células Cultivadas , Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/biossíntese , Humanos , Macrófagos/imunologia , Ligação Proteica , Multimerização Proteica , Receptor da Anafilatoxina C5a/química , Receptor da Anafilatoxina C5a/genética , Receptores de Quimiocinas/química , Receptores de Quimiocinas/genética , beta-Arrestina 2 , beta-Arrestinas
9.
Int J Med Microbiol ; 304(7): 877-93, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25082204

RESUMO

The distinctive and unique features of the avian and mammalian zoonotic pathogen Chlamydia (C.) psittaci include the fulminant course of clinical disease, the remarkably wide host range and the high proportion of latent infections that are not leading to overt disease. Current knowledge on associated diseases is rather poor, even in comparison to other chlamydial agents. In the present paper, we explain and summarize the major findings of a national research network that focused on the elucidation of host-pathogen interactions in vitro and in animal models of C. psittaci infection, with the objective of improving our understanding of genomics, pathology, pathophysiology, molecular pathogenesis and immunology, and conceiving new approaches to therapy. We discuss new findings on comparative genome analysis, the complexity of pathophysiological interactions and systemic consequences, local immune response, the role of the complement system and antigen presentation pathways in the general context of state-of-the-art knowledge on chlamydial infections in humans and animals and single out relevant research topics to fill remaining knowledge gaps on this important yet somewhat neglected pathogen.


Assuntos
Chlamydophila psittaci/genética , Chlamydophila psittaci/imunologia , Interações Hospedeiro-Patógeno , Patologia Clínica , Psitacose/imunologia , Psitacose/patologia , Animais , Chlamydophila psittaci/patogenicidade , Modelos Animais de Doenças , Genômica , Humanos , Psitacose/microbiologia
10.
Inflamm Res ; 63(3): 239-47, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24316866

RESUMO

OBJECTIVE: Expression and function of histamine H4-receptor, an immunomodulatory receptor involved in inflammatory diseases, on murine macrophages, which are vital for immunity, were investigated. MATERIALS AND METHODS: The expression pattern of histamine receptors on bone marrow-derived macrophages of BALB/c mice and on RAW 264.7 cells was studied at the mRNA level by reverse transcription polymerase chain reaction. The functional relevance of histamine receptors was investigated by analyzing histamine-induced chemotaxis and phagocytosis in the presence of histamine receptor antagonists mepyramine (histamine H1-receptor), famotidine (histamine H2-receptor), thioperamide (histamine H3/4-receptors) and JNJ7777120 (histamine H4-receptor). RESULTS: Both bone marrow-derived macrophages and RAW 264.7 cells express mRNA for histamine H1-receptor and histamine H4-receptor. Residual amounts of histamine H2-receptor mRNA are found in bone marrow-derived macrophages only. In both cellular models, histamine induced chemotaxis and phagocytic activity, which was reduced by thioperamide as well as by JNJ 7777120, but not by mepyramine or famotidine. CONCLUSION: In murine bone marrow-derived macrophages and RAW 264.7 macrophage-like cells histamine H4-receptor mediates chemotaxis and phagocytic activity.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Histamina/farmacologia , Macrófagos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Histamínicos/efeitos dos fármacos , Animais , Células da Medula Óssea/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/biossíntese , Receptores Histamínicos/biossíntese , Receptores Histamínicos H4
11.
J Allergy Clin Immunol ; 132(6): 1358-67, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23932072

RESUMO

BACKGROUND: Epidermal hyperproliferation resulting in acanthosis is an important clinical observation in patients with atopic dermatitis, and its underlying mechanisms are not completely understood. OBJECTIVE: Because increased levels of histamine are present in lesional skin, we investigated the effect of histamine, especially with regard to histamine 4 receptor (H4R) activation, on the proliferation of human and murine keratinocytes. METHODS: The expression of H4R on human and murine keratinocytes was detected by using real-time PCR. Keratinocyte proliferation was evaluated by using different in vitro cell proliferation assays, scratch assays, and measurement of the epidermal thickness of murine skin. RESULTS: We detected H4R mRNA on foreskin keratinocytes and on outer root sheath keratinocytes; H4R mRNA was more abundant in keratinocytes from patients with atopic dermatitis compared with those from nonatopic donors. Stimulation of foreskin keratinocytes, atopic dermatitis outer root sheath keratinocytes, and H4R-transfected HaCaT cells with histamine and H4R agonist resulted in an increase in proliferation, which was blocked with the H4R-specific antagonist JNJ7777120. Abdominal epidermis of H4R-deficient mice was significantly thinner, and the in vitro proliferation of keratinocytes derived from H4R-deficient mice was lower compared with that seen in control mice. Interestingly, we only detected H4R expression on murine keratinocytes after stimulation with LPS and peptidoglycan. CONCLUSION: H4R is highly expressed on keratinocytes from patients with atopic dermatitis, and its stimulation induces keratinocyte proliferation. This might represent a mechanism that contributes to the epidermal hyperplasia observed in patients with atopic dermatitis.


Assuntos
Dermatite Atópica/imunologia , Queratinócitos/imunologia , Receptores Acoplados a Proteínas G/biossíntese , Receptores Histamínicos/biossíntese , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Histamina/imunologia , Antagonistas dos Receptores Histamínicos/farmacologia , Humanos , Indóis/farmacologia , Queratinócitos/efeitos dos fármacos , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptidoglicano/imunologia , Piperazinas/farmacologia , Receptores Acoplados a Proteínas G/genética , Receptores Histamínicos/genética , Receptores Histamínicos H4
12.
Front Immunol ; 15: 1368040, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38562925

RESUMO

Background: Excessive inflammation, hemolysis, and accumulation of labile heme play an essential role in the pathophysiology of multi-organ dysfunction syndrome (MODS) in sepsis. Alpha1-antitrypsin (AAT), an acute phase protein with heme binding capacity, is one of the essential modulators of host responses to inflammation. In this study, we evaluate the putative protective effect of AAT against MODS and mortality in a mouse model of polymicrobial abdominal sepsis. Methods: Polymicrobial abdominal sepsis was induced in C57BL/6N mice by cecal ligation and puncture (CLP). Immediately after CLP surgery, mice were treated intraperitoneally with three different forms of human AAT-plasma-derived native (nAAT), oxidized nAAT (oxAAT), or recombinant AAT (recAAT)-or were injected with vehicle. Sham-operated mice served as controls. Mouse survival, bacterial load, kidney and liver function, immune cell profiles, cytokines/chemokines, and free (labile) heme levels were assessed. In parallel, in vitro experiments were carried out with resident peritoneal macrophages (MPMΦ) and mouse peritoneal mesothelial cells (MPMC). Results: All AAT preparations used reduced mortality in septic mice. Treatment with AAT significantly reduced plasma lactate dehydrogenase and s-creatinine levels, vascular leakage, and systemic inflammation. Specifically, AAT reduced intraperitoneal accumulation of free heme, production of cytokines/chemokines, and neutrophil infiltration into the peritoneal cavity compared to septic mice not treated with AAT. In vitro experiments performed using MPMC and primary MPMΦ confirmed that AAT not only significantly decreases lipopolysaccharide (LPS)-induced pro-inflammatory cell activation but also prevents the enhancement of cellular responses to LPS by free heme. In addition, AAT inhibits cell death caused by free heme in vitro. Conclusion: Data from the septic CLP mouse model suggest that intraperitoneal AAT treatment alone is sufficient to improve sepsis-associated organ dysfunctions, preserve endothelial barrier function, and reduce mortality, likely by preventing hyper-inflammatory responses and by neutralizing free heme.


Assuntos
Doenças Transmissíveis , Sepse , Humanos , Camundongos , Animais , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Citocinas/metabolismo , Inflamação/tratamento farmacológico , Quimiocinas , Fatores Imunológicos
13.
Infect Immun ; 81(9): 3366-74, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23817611

RESUMO

Chlamydia pneumoniae is associated with chronic inflammatory lung diseases like bronchial asthma and chronic obstructive pulmonary disease. The existence of a causal link between allergic airway disease and C. pneumoniae is controversial. A mouse model was used to address the question of whether preceding C. pneumoniae lung infection and recovery modifies the outcome of experimental allergic asthma after subsequent sensitization with house dust mite (HDM) allergen. After intranasal infection, BALB/c mice suffered from pneumonia characterized by an increased clinical score, reduction of body weight, histopathology, and a bacterial load in the lungs. After 4 weeks, when infection had almost resolved clinically, HDM allergen sensitization was performed for another 4 weeks. Subsequently, mice were subjected to a methacholine hyperresponsiveness test and sacrificed for further analyses. As expected, after 8 weeks, C. pneumoniae-specific antibodies were detectable only in infected mice and the titer was significantly higher in the C. pneumoniae/HDM allergen-treated group than in the C. pneumoniae/NaCl group. Intriguingly, airway hyperresponsiveness and eosinophilia in bronchoalveolar lavage fluid were significantly lower in the C. pneumoniae/HDM allergen-treated group than in the mock/HDM allergen-treated group. We did observe a relationship between experimental asthma and chlamydial infection. Our results demonstrate an influence of sensitization to HDM allergen on the development of a humoral antibacterial response. However, our model demonstrates no increase in the severity of experimental asthma to HDM allergen as a physiological allergen after clinically resolved severe chlamydial lung infection. Our results rather suggest that allergic airway disease and concomitant cellular changes in mice are decreased following C. pneumoniae lung infection in this setting.


Assuntos
Alérgenos/imunologia , Chlamydophila pneumoniae/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Pyroglyphidae/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/patologia , Animais , Asma/imunologia , Asma/microbiologia , Asma/patologia , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Infecções por Chlamydophila/imunologia , Infecções por Chlamydophila/microbiologia , Infecções por Chlamydophila/patologia , Eosinofilia/imunologia , Eosinofilia/microbiologia , Eosinofilia/patologia , Feminino , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/imunologia , Pneumonia/microbiologia , Pneumonia/patologia , Infecções Respiratórias/microbiologia
14.
J Immunol ; 186(5): 3066-75, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21263075

RESUMO

Complement activation represents a crucial innate defense mechanism to invading microorganisms, but there is an eminent lack of understanding of the separate contribution of the different complement activation pathways to the host response during sepsis. We therefore investigated different innate host immune responses during cecal ligation and puncture (CLP)-induced sepsis in mice lacking either the alternative (fD(-/-)) or classical (C1q(-/-)) complement activation pathway. Both knockout mice strains showed a significantly reduced survival and increased organ dysfunction when compared with control mice. Surprisingly, fD(-/-) mice demonstrated a compensated bacterial clearance capacity as control mice at 6 h post CLP, whereas C1q(-/-) mice were already overwhelmed by bacterial growth at this time point. Interestingly, at 24 h after CLP, fD(-/-) mice failed to clear bacteria in a way comparable to control mice. However, both knockout mice strains showed compromised C3 cleavage during sepsis. Investigating potential causes for this discrepancy, we were able to demonstrate that despite normal bacterial clearance capacity early during the onset of sepsis, fD(-/-) mice displayed increased inflammatory cytokine generation and neutrophil recruitment into lungs and blood when compared with both control- and C1q(-/-) mice, indicating a potential loss of control over these immune responses. Further in vitro experiments revealed a strongly increased Nf-κB activation capacity in isolated neutrophils from fD(-/-) mice, supporting this hypothesis. Our results provide evidence for the new concept that the alternative complement activation pathway exerts a distinctly different contribution to the innate host response during sepsis when compared with the classical pathway.


Assuntos
Ativação do Complemento/imunologia , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/imunologia , Choque Séptico/imunologia , Animais , Carga Bacteriana/imunologia , Ceco , Ativação do Complemento/genética , Complemento C1q/deficiência , Complemento C1q/genética , Fator D do Complemento/deficiência , Fator D do Complemento/genética , Via Alternativa do Complemento/genética , Via Clássica do Complemento/genética , Imunidade Inata/genética , Rim/imunologia , Rim/microbiologia , Rim/fisiopatologia , Ligadura , Fígado/imunologia , Fígado/microbiologia , Fígado/fisiopatologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Punções , Choque Séptico/genética , Choque Séptico/mortalidade , Análise de Sobrevida
15.
Front Immunol ; 14: 1072142, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36761727

RESUMO

Infections with influenza A viruses (IAV) cause seasonal epidemics and global pandemics. The majority of these infections remain asymptomatic, especially among children below five years of age. Importantly, this is a time, when immunological imprinting takes place. Whether early-life infections with IAV affect the development of antimicrobial immunity is unknown. Using a preclinical mouse model, we demonstrate here that silent neonatal influenza infections have a remote beneficial impact on the later control of systemic juvenile-onset and adult-onset infections with an unrelated pathogen, Staphylococcus aureus, due to improved pathogen clearance and clinical resolution. Strategic vaccination with a live attenuated IAV vaccine elicited a similar protection phenotype. Mechanistically, the IAV priming effect primarily targets antimicrobial functions of the developing innate immune system including increased antimicrobial plasma activity and enhanced phagocyte functions and antigen-presenting properties at mucosal sites. Our results suggest a long-term benefit from an exposure to IAV during the neonatal phase, which might be exploited by strategic vaccination against influenza early in life to enforce the host's resistance to later bacterial infections.


Assuntos
Anti-Infecciosos , Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Animais , Camundongos , Humanos
16.
J Immunol ; 185(9): 5628-36, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20870944

RESUMO

The complement system as well as the coagulation system has fundamental clinical implications in the context of life-threatening tissue injury and inflammation. Associations between both cascades have been proposed, but the precise molecular mechanisms remain unknown. The current study reports multiple links for various factors of the coagulation and fibrinolysis cascades with the central complement components C3 and C5 in vitro and ex vivo. Thrombin, human coagulation factors (F) XIa, Xa, and IXa, and plasmin were all found to effectively cleave C3 and C5. Mass spectrometric analyses identified the cleavage products as C3a and C5a, displaying identical molecular weights as the native anaphylatoxins C3a and C5a. Cleavage products also exhibited robust chemoattraction of human mast cells and neutrophils, respectively. Enzymatic activity for C3 cleavage by the investigated clotting and fibrinolysis factors is defined in the following order: FXa > plasmin > thrombin > FIXa > FXIa > control. Furthermore, FXa-induced cleavage of C3 was significantly suppressed in the presence of the selective FXa inhibitors fondaparinux and enoxaparin in a concentration-dependent manner. Addition of FXa to human serum or plasma activated complement ex vivo, represented by the generation of C3a, C5a, and the terminal complement complex, and decreased complement hemolytic serum activity that defines exact serum concentration that results in complement-mediated lysis of 50% of sensitized sheep erythrocytes. Furthermore, in plasma from patients with multiple injuries (n = 12), a very early appearance and correlation of coagulation (thrombin-antithrombin complexes) and the complement activation product C5a was found. The present data suggest that coagulation/fibrinolysis proteases may act as natural C3 and C5 convertases, generating biologically active anaphylatoxins, linking both cascades via multiple direct interactions in terms of a complex serine protease system.


Assuntos
Anafilatoxinas/metabolismo , Coagulação Sanguínea/fisiologia , Ativação do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Serina Proteases/metabolismo , Adulto , Idoso , Western Blotting , Quimiotaxia de Leucócito/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Mastócitos/imunologia , Mastócitos/metabolismo , Pessoa de Meia-Idade , Neutrófilos/imunologia , Neutrófilos/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Adulto Jovem
17.
Vaccines (Basel) ; 9(8)2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34451996

RESUMO

Chlamydia trachomatis causes most bacterial sexually transmitted diseases worldwide. Different major outer membrane proteins (MOMPs) define various serovars of this intracellular pathogen: In women, D to L3 can cause urethritis, cervicitis, salpingitis, and oophoritis, and, thus, infertility. Protective immunity might be serovar-specific since chlamydial infection does not appear to induce an effective acquired immunity and reinfections occur. A better understanding of induced cross-serovar protection is essential for the selection of suitable antigens in vaccine development. In our mouse lung infection screening model, we evaluated the urogenital serovars D, E, and L2 in this regard. Seven weeks after primary infection or mock-infection, respectively, mice were infected a second time with the identical or one of the other serovars. Body weight and clinical score were monitored for 7 days. Near the peak of the second lung infection, bacterial load, myeloperoxidase, IFN-γ, and TNF-α in lung homogenate, as well as chlamydia-specific IgG levels in blood were determined. Surprisingly, compared with mice that were infected then for the first time, almost independent of the serovar combination used, all acquired parameters of disease were similarly diminished. Our reinfection study suggests that efficient cross-serovar protection could be achieved by a vaccine combining chlamydial antigens that do not include nonconserved MOMP regions.

18.
Front Immunol ; 12: 626627, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33746963

RESUMO

Recent advances in complement research have revolutionized our understanding of its role in immune responses. The immunomodulatory features of complement in infections by intracellular pathogens, e.g., viruses, are attracting increasing attention. Thereby, local production and activation of complement by myeloid-derived cells seem to be crucial. We could recently show that C3, a key player of the complement cascade, is required for effective defense against the intracellular bacterium Chlamydia psittaci. Avian zoonotic strains of this pathogen cause life-threatening pneumonia with systemic spread in humans; closely related non-avian strains are responsible for less severe diseases of domestic animals with economic loss. To clarify how far myeloid- and non-myeloid cell-derived complement contributes to immune response and resulting protection against C. psittaci, adoptive bone marrow transfer experiments focusing on C3 were combined with challenge experiments using a non-avian (BSL 2) strain of this intracellular bacterium. Surprisingly, our data prove that for C. psittaci-induced pneumonia in mice, non-myeloid-derived, circulating/systemic C3 has a leading role in protection, in particular on the development of pathogen-specific T- and B- cell responses. In contrast, myeloid-derived and most likely locally produced C3 plays only a minor, mainly fine-tuning role. The work we present here describes authentic, although less pronounced, antigen directed immune responses.


Assuntos
Imunidade Adaptativa , Infecções por Chlamydia/microbiologia , Chlamydophila psittaci/patogenicidade , Complemento C3/metabolismo , Pulmão/microbiologia , Pneumonia Bacteriana/microbiologia , Transferência Adotiva , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/microbiologia , Transplante de Medula Óssea , Células Cultivadas , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/metabolismo , Chlamydophila psittaci/imunologia , Complemento C3/genética , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/microbiologia , Quimeras de Transplante
19.
Front Immunol ; 12: 580594, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33767691

RESUMO

The zoonotic intracellular bacterium Chlamydia psittaci causes life-threatening pneumonia in humans. During mouse lung infection, complement factor C3 and the anaphylatoxin C3a augment protection against C. psittaci by a so far unknown mechanism. To clarify how complement contributes to the early, innate and the late, specific immune response and resulting protection, this study addresses the amount of C3, the timing when its presence is required as well as the anaphylatoxin receptor(s) mediating its effects and the complement-dependent migration of dendritic cells. Challenge experiments with C. psittaci on various complement KO mice were combined with transient decomplementation by pharmacological treatment, as well as the analysis of in vivo dendritic cells migration. Our findings reveal that a plasma concentration of C3 close to wildtype levels was required to achieve full protection. The diminished levels of C3 of heterozygote C3+/- mice permitted already relative effective protection and improved survival as compared to C3-/- mice, but overall recovery of these animals was delayed. Complement was in particular required during the first days of infection. However, additionally, it seems to support protection at later stages. Migration of CD103+ dendritic cells from the infected lung to the draining lymph node-as prerequisite of antigen presentation-depended on C3 and C3aR and/or C5aR. Our results provide unique mechanistic insight in various aspects of complement-dependent immune responses under almost identical, rather physiological experimental conditions. Our study contributes to an improved understanding of the role of complement, and C3a in particular, in infections by intracellular bacteria.


Assuntos
Movimento Celular/imunologia , Infecções por Chlamydiaceae/imunologia , Chlamydophila psittaci/imunologia , Complemento C3a/imunologia , Células Dendríticas/imunologia , Pulmão/imunologia , Anafilatoxinas/imunologia , Anafilatoxinas/metabolismo , Animais , Linhagem Celular , Infecções por Chlamydiaceae/metabolismo , Infecções por Chlamydiaceae/microbiologia , Chlamydophila psittaci/fisiologia , Ativação do Complemento/imunologia , Complemento C3a/genética , Complemento C3a/metabolismo , Células Dendríticas/citologia , Células Dendríticas/microbiologia , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Complemento/genética , Receptores de Complemento/imunologia , Receptores de Complemento/metabolismo , Transdução de Sinais/imunologia , Análise de Sobrevida
20.
Vaccines (Basel) ; 9(6)2021 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-34204170

RESUMO

Chlamydia trachomatis is the most frequent sexually-transmitted disease-causing bacterium. Urogenital serovars of this intracellular pathogen lead to urethritis and cervicitis. Ascending infections result in pelvic inflammatory disease, salpingitis, and oophoritis. One of 200 urogenital infections leads to tubal infertility. Serovars A-C cause trachoma with visual impairment. There is an urgent need for a vaccine. We characterized a new five-component subunit vaccine in a mouse vaccination-lung challenge infection model. Four recombinant Pmp family-members and Ctad1 from C. trachomatis serovar E, all of which participate in adhesion and binding of chlamydial elementary bodies to host cells, were combined with the mucosal adjuvant cyclic-di-adenosine monophosphate. Intranasal application led to a high degree of cross-serovar protection against urogenital and ocular strains of C. trachomatis, which lasted at least five months. Critical evaluated parameters were body weight, clinical score, chlamydial load, a granulocyte marker and the cytokines IFN-γ/TNF-α in lung homogenate. Vaccine antigen-specific antibodies and a mixed Th1/Th2/Th17 T cell response with multi-functional CD4+ and CD8+ T cells correlate with protection. However, serum-transfer did not protect the recipients suggesting that circulating antibodies play only a minor role. In the long run, our new vaccine might help to prevent the feared consequences of human C. trachomatis infections.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA