Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
Sci Rep ; 11(1): 14961, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294757

RESUMO

Influenza and other respiratory viruses present a significant threat to public health, national security, and the world economy, and can lead to the emergence of global pandemics such as from COVID-19. A barrier to the development of effective therapeutics is the absence of a robust and predictive preclinical model, with most studies relying on a combination of in vitro screening with immortalized cell lines and low-throughput animal models. Here, we integrate human primary airway epithelial cells into a custom-engineered 96-device platform (PREDICT96-ALI) in which tissues are cultured in an array of microchannel-based culture chambers at an air-liquid interface, in a configuration compatible with high resolution in-situ imaging and real-time sensing. We apply this platform to influenza A virus and coronavirus infections, evaluating viral infection kinetics and antiviral agent dosing across multiple strains and donor populations of human primary cells. Human coronaviruses HCoV-NL63 and SARS-CoV-2 enter host cells via ACE2 and utilize the protease TMPRSS2 for spike protein priming, and we confirm their expression, demonstrate infection across a range of multiplicities of infection, and evaluate the efficacy of camostat mesylate, a known inhibitor of HCoV-NL63 infection. This new capability can be used to address a major gap in the rapid assessment of therapeutic efficacy of small molecules and antiviral agents against influenza and other respiratory viruses including coronaviruses.


Assuntos
Antivirais/farmacologia , Infecções por Coronavirus/virologia , Influenza Humana/virologia , Testes de Sensibilidade Microbiana/instrumentação , Técnicas Analíticas Microfluídicas/instrumentação , Mucosa Respiratória/citologia , Brônquios/citologia , Brônquios/virologia , COVID-19/virologia , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Coronavirus/efeitos dos fármacos , Infecções por Coronavirus/tratamento farmacológico , Desenho de Equipamento , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Mucosa Respiratória/virologia , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/virologia , SARS-CoV-2/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
2.
J Hosp Infect ; 96(3): 244-249, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28454768

RESUMO

BACKGROUND: Contact precautions are a widely accepted strategy to reduce in-hospital transmission of meticillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). However, these practices may have unintended deleterious effects on patients. AIM: To evaluate the effect of a modification in hospital-wide contact precaution practices on emergency department (ED) admission times. METHODS: During the study period, the hospital changed its contact precaution policy from requiring contact precautions for all patients with a history of MRSA or VRE to only those who presented with clinical conditions likely to contaminate the environment with pathogens. An interrupted time series analysis of ED admission times for adults for one year preceding and one year following this change was performed at a two-campus hospital. The main outcome was admission time, defined as time from decision to admit to arrival in an inpatient bed, for patients with MRSA or VRE compared with all other patients. The in-hospital MRSA and VRE acquisition rates were evaluated over the same period and have been published previously. FINDINGS: At one campus, admission time decreased immediately by 161min for MRSA patients (P=0.008) and 135min for VRE patients (P=0.003), and both continued to decrease over the duration of the study. There was no significant change in admission time at the second campus. CONCLUSIONS: Modifying contact precaution requirements for MRSA and VRE may be associated with improved ED admission time without significantly altering in-hospital MRSA and VRE acquisition.


Assuntos
Infecção Hospitalar/prevenção & controle , Medicina de Emergência/métodos , Infecções por Bactérias Gram-Positivas/diagnóstico , Controle de Infecções/métodos , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Admissão do Paciente , Enterococos Resistentes à Vancomicina/isolamento & purificação , Adulto , Portador Sadio/diagnóstico , Serviço Hospitalar de Emergência , Hospitais , Humanos , Política Organizacional , Estudos Retrospectivos , Fatores de Tempo
3.
Am J Transplant ; 13(4): 1047-1054, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23356386

RESUMO

Rapid allograft infection complicates liver transplantation (LT) in patients with hepatitis C virus (HCV). Pegylated interferon-α and ribavirin therapy after LT has significant toxicity and limited efficacy. The effect of a human monoclonal antibody targeting the HCV E2 glycoprotein (MBL-HCV1) on viral clearance was examined in a randomized, double-blind, placebo-controlled pilot study in patients infected with HCV genotype 1a undergoing LT. Subjects received 11 infusions of 50 mg/kg MBL-HCV1 (n=6) or placebo (n=5) intravenously with three infusions on day of transplant, a single infusion on days 1 through 7 and one infusion on day 14 after LT. MBL-HCV1 was well-tolerated and reduced viral load for a period ranging from 7 to 28 days. Median change in viral load (log10 IU/mL) from baseline was significantly greater (p=0.02) for the antibody-treated group (range -3.07 to -3.34) compared to placebo group (range -0.331 to -1.01) on days 3 through 6 posttransplant. MBL-HCV1 treatment significantly delayed median time to viral rebound compared to placebo treatment (18.7 days vs. 2.4 days, p<0.001). As with other HCV monotherapies, antibody-treated subjects had resistance-associated variants at the time of viral rebound. A combination study of MBL-HCV1 with a direct-acting antiviral is underway.


Assuntos
Anticorpos Monoclonais/farmacologia , Hepacivirus/fisiologia , Hepatite C/tratamento farmacológico , Transplante de Fígado , Idoso , Biópsia , Método Duplo-Cego , Feminino , Genótipo , Hepatite C/virologia , Humanos , Fígado/patologia , Masculino , Pessoa de Meia-Idade , Projetos Piloto , RNA Viral/análise , Fatores de Tempo , Proteínas do Envelope Viral/imunologia
4.
Clin Exp Immunol ; 140(2): 230-40, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15807846

RESUMO

Complement receptor 1 (CR1) on the surface of human erythrocytes facilitates intravascular clearance of complement-opsonized pathogens. The need for complement activation can be circumvented by directly coupling the organism to CR1 using a bispecific monoclonal antibody heteropolymer (HP). Lack of a functional homologue to CR1 on mouse erythrocytes has made it difficult to study HP-dependent clearance of pathogens in small animals. We have developed a transgenic mouse that expresses human CR1 on erythrocytes. CR1 antigen is of appropriate size and in a clustered distribution as confirmed by immunoblotting and fluorescence microscopy, respectively. HP that immobilized bacteriophage PhiX174 prototype pathogen to erythrocyte CR1 of the transgenic mice increased the rate of clearance of the virus compared with HP that bound bacteriophage, but not CR1. This transgenic mouse model will allow evaluation of different HPs for their in vivo efficacy and potential as human therapeutics.


Assuntos
Anticorpos Biespecíficos/imunologia , Patógenos Transmitidos pelo Sangue , Modelos Animais de Doenças , Receptores de Complemento/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Bacteriófago phi X 174/imunologia , Relação Dose-Resposta Imunológica , Eritrócitos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Papio
5.
J Virol ; 75(22): 10730-7, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602714

RESUMO

The mammalian Toll-like receptor 4, TLR4, is an important component in the innate immune response to gram-negative bacterial infection. The role of TLR4 in antiviral immunity has been largely unexplored. In this study, the in vivo immune responses to respiratory syncytial virus (RSV) and influenza virus infection were examined in TLR4-deficient (C57BL/10ScNCr) and TLR4-expressing (C57BL/10Sn) mice. TLR4-deficient mice challenged with RSV, but not influenza virus, exhibited impaired natural killer (NK) cell and CD14(+) cell pulmonary trafficking, deficient NK cell function, impaired interleukin-12 expression, and impaired virus clearance compared to mice expressing TLR4. These findings suggest that Toll signaling pathways have an important role in innate immunity to RSV.


Assuntos
Proteínas de Drosophila , Glicoproteínas de Membrana/fisiologia , Receptores de Superfície Celular/fisiologia , Infecções por Vírus Respiratório Sincicial/imunologia , Animais , Citotoxicidade Imunológica , Feminino , Imunidade Inata , Interleucina-12/fisiologia , Interleucina-18/fisiologia , Células Matadoras Naturais/imunologia , Receptores de Lipopolissacarídeos/análise , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Receptor 4 Toll-Like , Receptores Toll-Like
6.
Curr Opin Oncol ; 13(5): 349-53, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11555711

RESUMO

Epstein-Barr virus (EBV), a human herpes virus, is associated with a variety of malignancies. In vitro, it is able to transform B cells, which will grow as lymphoblastoid cell lines in the absence of T cells. Patients with a variety of immunodeficiency diseases are subject to the development of B-cell lymphomas that express viral antigens on their cell surface. Development of EBV-associated B-cell lymphomas is seen in solid organ transplant and bone marrow transplant recipients receiving immunosuppressive therapy. Transfer of mature T cells from EBV-immune marrow donors has been demonstrated to be effective in controlling these EBV-associated B-cell tumors. Recently the demonstration that EBV transcripts are found in other lymphomas (including Hodgkin disease cells) has led to the suggestion that transfer of EBV-specific T cells may also be effective in managing these tumors. Current research involves optimizing methods to expand cells that recognize the EBV antigens expressed in the lymphoma cells.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4/imunologia , Linfoma de Células B/virologia , Linfócitos T/virologia , Antígenos Virais , Humanos , Hospedeiro Imunocomprometido , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Transplante de Órgãos/efeitos adversos , Linfócitos T/imunologia
7.
Infect Immun ; 68(12): 6650-5, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11083777

RESUMO

Abscesses are a classic host response to infection by many pathogenic bacteria. The immunopathogenesis of this tissue response to infection has not been fully elucidated. Previous studies have suggested that T cells are involved in the pathologic process, but the role of these cells remains unclear. To delineate the mechanism by which T cells mediate abscess formation associated with intra-abdominal sepsis, the role of T-cell activation and the contribution of antigen-presenting cells via CD28-B7 costimulation were investigated. T cells activated in vitro by zwitterionic bacterial polysaccharides (Zps) known to induce abscess formation required CD28-B7 costimulation and, when adoptively transferred to the peritoneal cavity of naïve rats, promoted abscess formation. Blockade of T-cell activation via the CD28-B7 pathway in animals with CTLA4Ig prevented abscess formation following challenge with different bacterial pathogens, including Staphylococcus aureus, Bacteroides fragilis, and a combination of Enterococcus faecium and Bacteroides distasonis. In contrast, these animals had an increased abscess rate following in vivo T-cell activation via CD28 signaling. Abscess formation in vivo and T-cell activation in vitro required costimulation by B7-2 but not B7-1. These results demonstrate that abscess formation by pathogenic bacteria is under the control of a common effector mechanism that requires T-cell activation via the CD28-B7-2 pathway.


Assuntos
Abscesso/etiologia , Antígenos CD/fisiologia , Antígenos CD28/fisiologia , Linfócitos T CD4-Positivos/imunologia , Imunoconjugados , Ativação Linfocitária , Glicoproteínas de Membrana/fisiologia , Abatacepte , Animais , Antígenos de Diferenciação/farmacologia , Antígeno B7-1/fisiologia , Antígeno B7-2 , Antígeno CTLA-4 , Humanos , Masculino , Ratos , Ratos Wistar
8.
Virology ; 271(1): 99-108, 2000 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10814575

RESUMO

Group B coxsackieviruses are etiologically linked to many human diseases, and cell surface receptors are postulated to play an important role in mediating their pathogenesis. The coxsackievirus adenovirus receptor (CAR) has been shown to function as a receptor for selected strains of coxsackievirus group B (CVB) serotypes 3, 4, and 5 and is postulated to serve as a receptor for all six serotypes. In this study, we demonstrate that CAR can serve as a receptor for laboratory reference strains and clinical isolates of all six CVB serotypes. Infection of CHO cells expressing human CAR results in a 1000-fold increase in CVB progeny virus titer compared to mock transfected cells. CAR was shown to be a functional receptor for swine vesicular disease virus (SVDV), as CHO-CAR cells but not CHO mock transfected controls were susceptible to SVDV infection, produced progeny SVDV, and developed cytopathic effects. Moreover, SVDV infection could be specifically blocked by monoclonal antibody to CAR (RmcB). SVDV infection of HeLa cells was also inhibited by an anti-CD55 MAb, suggesting that this virus, like some CVB, may interact with CD55 (decay accelerating factor) in addition to CAR. Finally, pretreatment of CVB or SVDV with soluble CAR effectively blocks virus infection of HeLa cell monolayers.


Assuntos
Enterovirus Humano B/classificação , Receptores Virais/fisiologia , Doença Vesicular Suína/virologia , Animais , Antígenos CD55/metabolismo , Células CHO , Chlorocebus aethiops , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Cricetinae , Efeito Citopatogênico Viral , Células HeLa , Humanos , Reação em Cadeia da Polimerase , Sorotipagem , Suínos , Transfecção , Células Vero
9.
J Biol Chem ; 275(10): 6733-40, 2000 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-10702228

RESUMO

Immunologic paradigms classify bacterial polysaccharides as T cell-independent antigens. However, these models fail to explain how zwitterionic polysaccharides (Zps) confer protection against intraabdominal abscess formation in a T cell-dependent manner. Here, we demonstrate that Zps elicit a potent CD4+ T cell response in vitro that requires available major histocompatibility complex class II molecules on antigen-presenting cells. Specific chemical modifications to Zps show that: 1) the activity is specific for carbohydrate structure, and 2) the proliferative response depends upon free amino and carboxyl groups on the repeating units of these polysaccharides. Peptides synthesized to mimic the zwitterionic charge motif associated with Zps also exhibited these biologic properties. Lysine-aspartic acid (KD) peptides with more than 15 repeating units stimulated CD4+ T cells in vitro and conferred protection against abscesses induced by bacteria such as Bacteroides fragilis and Staphylococcus aureus. Evidence for the biologic importance of T cell activation by these zwitterionic polymers was provided when human CD4+ T cells stimulated with these molecules in vitro and adoptively transferred to rats in vivo conferred protection against intraabdominal abscesses induced by viable bacterial challenge. These studies demonstrate that bacterial polysaccharides with a distinct charge motif activate T cells and that this activity confers immunity to a distinct pathologic response to bacterial infection.


Assuntos
Abscesso/prevenção & controle , Ativação Linfocitária/efeitos dos fármacos , Polissacarídeos Bacterianos/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Bacteroides fragilis/imunologia , Técnicas In Vitro , Masculino , Peptídeos/farmacologia , Ratos , Ratos Endogâmicos Lew , Streptococcus pneumoniae/imunologia , Relação Estrutura-Atividade , Linfócitos T/imunologia
10.
Nat Med ; 6(4): 435-42, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10742151

RESUMO

Here, we demonstrate a previously unknown function for the 70-kDa heat-shock protein (HSP70) as a cytokine. HSP70 bound with high affinity to the plasma membrane, elicited a rapid intracellular calcium flux, activated nuclear factor (NF)-kappaB and upregulated the expression of pro-inflammatory cytokines tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta and IL-6 in human monocytes. Furthermore, two different signal transduction pathways were activated by exogenous HSP70: one dependent on CD14 and intracellular calcium, which resulted in increased IL-1beta, IL-6 and TNF-alpha; and the other independent of CD14 but dependent on intracellular calcium, which resulted in an increase in TNF-alpha but not IL-1beta or IL-6. These findings indicate that CD14 is a co-receptor for HSP70-mediated signaling in human monocytes and are indicative of an previously unrecognized function for HSP70 as an extracellular protein with regulatory effects on human monocytes, having a dual role as chaperone and cytokine.


Assuntos
Proteínas de Choque Térmico HSP70/imunologia , Proteínas I-kappa B , Interleucina-1/biossíntese , Interleucina-6/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Cálcio/imunologia , Células Cultivadas , Citocinas/imunologia , Citocinas/fisiologia , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Choque Térmico HSP70/farmacologia , Humanos , Receptores de Lipopolissacarídeos/imunologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Células Tumorais Cultivadas
11.
J Clin Invest ; 105(4): 497-504, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10683379

RESUMO

Lipopolysaccharide (LPS) is the main inducer of shock and death in Gram-negative sepsis. Recent evidence suggests that LPS-induced signal transduction begins with CD14-mediated activation of 1 or more Toll-like receptors (TLRs). The lipid A analogues lipid IVa and Rhodobacter sphaeroides lipid A (RSLA) exhibit an uncommon species-specific pharmacology. Both compounds inhibit the effects of LPS in human cells but display LPS-mimetic activity in hamster cells. We transfected human TLR4 or human TLR2 into hamster fibroblasts to determine if either of these LPS signal transducers is responsible for the species-specific pharmacology. RSLA and lipid IVa strongly induced NF-kappaB activity and IL-6 release in Chinese hamster ovary fibroblasts expressing CD14 (CHO/CD14), but these compounds antagonized LPS antagonists in CHO/CD14 fibroblasts that overexpressed human TLR4. No such antagonism occurred in cells overexpressing human TLR2. We cloned TLR4 from hamster macrophages and found that human THP-1 cells expressing the hamster TLR4 responded to lipid IVa as an LPS mimetic, as if they were hamster in origin. Hence, cells heterologously overexpressing TLR4 from different species acquired a pharmacological phenotype with respect to recognition of lipid A substructures that corresponded to the species from which the TLR4 transgene originated. These data suggest that TLR4 is the central lipid A-recognition protein in the LPS receptor complex.


Assuntos
Proteínas de Drosophila , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Células CHO , Cricetinae , Glicolipídeos/metabolismo , Humanos , Ligantes , Lipídeo A/análogos & derivados , Lipídeo A/metabolismo , Receptores de Lipopolissacarídeos/genética , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/citologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Mimetismo Molecular , Receptores de Superfície Celular/genética , Proteínas Recombinantes/metabolismo , Rhodobacter sphaeroides , Transdução de Sinais , Especificidade da Espécie , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like
12.
J Immunol ; 164(2): 719-24, 2000 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-10623815

RESUMO

The large-molecular-sized zwitterionic capsular polysaccharide of the anaerobe Bacteroides fragilis NCTC 9343, designated polysaccharide (PS) A, stimulates T cell proliferation in vitro and induces T cell-dependent protection against abscess formation in vivo. In the present study, we utilized a modification of a recently developed ozonolytic method for depolymerizing polysaccharides to examine the influence of the molecular size of PS A on cell-mediated immunity. Ozonolysis successfully depolymerized PS A into structurally intact fragments. PS A with average molecular sizes of 129.0 (native), 77.8, 46.9, and 17.1 kDa stimulated CD4+-cell proliferation in vitro to the same degree, whereas the 5.0-kDa fragment was much less stimulatory than the control 129.0-kDa PS A. Rats treated with 129.0-kDa, 46.9-kDa, and 17.1-kDa PS A molecules, but not those treated with the 5.0-kDa molecule, were protected against intraabdominal abscesses induced by challenge with viable B. fragilis. These results demonstrate that a zwitterionic polysaccharide as small as 22 repeating units (88 monosaccharides) elicits a T cell-dependent immune response. These findings clearly distinguish zwitterionic T cell-dependent polysaccharides from T cell-independent polysaccharides and give evidence of the existence of a novel mechanism for a polysaccharide-induced immune response.


Assuntos
Ativação Linfocitária/imunologia , Polissacarídeos Bacterianos/química , Polissacarídeos Bacterianos/imunologia , Abscesso Abdominal/imunologia , Abscesso Abdominal/prevenção & controle , Animais , Infecções por Bacteroides/imunologia , Infecções por Bacteroides/prevenção & controle , Bacteroides fragilis/imunologia , Soluções Tampão , Sequência de Carboidratos , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Humanos , Íons , Masculino , Dados de Sequência Molecular , Peso Molecular , Polissacarídeos Bacterianos/metabolismo , Ratos , Ratos Wistar
13.
J Immunol ; 164(1): 13-7, 2000 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-10604986

RESUMO

Heat shock proteins (HSP), highly conserved across species, are generally viewed as intracellular proteins thought to serve protective functions against infection and cellular stress. Recently, we have reported the surprising finding that human and chlamydial HSP60, both present in human atheroma, can activate vascular cells and macrophages. However, the transmembrane signaling pathways by which extracellular HSP60 may activate cells remains unclear. CD14, the monocyte receptor for LPS, binds numerous microbial products and can mediate activation of monocytes/macrophages and endothelial cells, thus promoting the innate immune response. We show here that human HSP60 activates human PBMC and monocyte-derived macrophages through CD14 signaling and p38 mitogen-activated protein kinase, sharing this pathway with bacterial LPS. These findings provide further insight into the molecular mechanisms by which extracellular HSP may participate in atherosclerosis and other inflammatory disorders by activating the innate immune system.


Assuntos
Chaperonina 60/fisiologia , Receptores de Lipopolissacarídeos/fisiologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Animais , Células CHO , Células Cultivadas , Chlamydia/imunologia , Cricetinae , Citocinas/biossíntese , Endotoxinas/fisiologia , Ativação Enzimática/imunologia , Genes Reporter/imunologia , Humanos , Imunidade Inata , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monócitos/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
14.
Nat Immunol ; 1(5): 398-401, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11062499

RESUMO

The innate immune system contributes to the earliest phase of the host defense against foreign organisms and has both soluble and cellular pattern recognition receptors for microbial products. Two important members of this receptor group, CD14 and the Toll-like receptor (TLR) pattern recognition receptors, are essential for the innate immune response to components of Gram-negative and Gram-positive bacteria, mycobacteria, spirochetes and yeast. We now find that these receptors function in an antiviral response as well. The innate immune response to the fusion protein of an important respiratory pathogen of humans, respiratory syncytial virus (RSV), was mediated by TLR4 and CD14. RSV persisted longer in the lungs of infected TLR4-deficient mice compared to normal mice. Thus, a common receptor activation pathway can initiate innate immune responses to both bacterial and viral pathogens.


Assuntos
Proteínas de Drosophila , Receptores de Lipopolissacarídeos/imunologia , Glicoproteínas de Membrana/imunologia , Receptores de Superfície Celular/imunologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/patogenicidade , Animais , Anticorpos Monoclonais/farmacologia , Citocinas/biossíntese , Humanos , Técnicas In Vitro , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/imunologia , Receptores de Lipopolissacarídeos/genética , Pulmão/imunologia , Pulmão/virologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Receptor 4 Toll-Like , Receptores Toll-Like , Proteínas Virais de Fusão/imunologia
15.
J Biol Chem ; 274(47): 33419-25, 1999 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-10559223

RESUMO

Toll-like receptors (TLRs) 2 and 4 are signal transducers for lipopolysaccharide, the major proinflammatory constituent in the outer membrane of Gram-negative bacteria. We observed that membrane lipoproteins/lipopeptides from Borrelia burgdorferi, Treponema pallidum, and Mycoplasma fermentans activated cells heterologously expressing TLR2 but not those expressing TLR1 or TLR4. These TLR2-expressing cells were also stimulated by living motile B. burgdorferi, suggesting that TLR2 recognition of lipoproteins is relevant to natural Borrelia infection. Importantly, a TLR2 antibody inhibited bacterial lipoprotein/lipopeptide-induced tumor necrosis factor release from human peripheral blood mononuclear cells, and TLR2-null Chinese hamster macrophages were insensitive to lipoprotein/lipopeptide challenge. The data suggest a role for the native protein in cellular activation by these ligands. In addition, TLR2-dependent responses were seen using whole Mycobacterium avium and Staphylococcus aureus, demonstrating that this receptor can function as a signal transducer for a wide spectrum of bacterial products. We conclude that diverse pathogens activate cells through TLR2 and propose that this molecule is a central pattern recognition receptor in host immune responses to microbial invasion.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Drosophila , Lipoproteínas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Grupo Borrelia Burgdorferi/metabolismo , Células CHO , Cricetinae , Humanos , Mycobacterium avium/metabolismo , Mycoplasma fermentans/metabolismo , Ligação Proteica , Receptor 1 Toll-Like , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like , Treponema pallidum/metabolismo
16.
J Acquir Immune Defic Syndr ; 22(1): 1-9, 1999 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10534141

RESUMO

Although nitric oxide (NO) production is increased in HIV-1-infected patients, and NO is known to inhibit the replication of several viruses, very little is known about the effects of NO on HIV-1 replication. In the present studies, we find that S-nitrosothiols (RSNOs), a class of NO donor compounds present in the human circulatory system, inhibit HIV-1 replication in acutely infected human peripheral blood mononuclear cells (PBMCs) and have an additive inhibitory effect on HIV-1 replication in combination with 3'-azido-3'-deoxythymidylate (AZT). RSNOs inhibit HIV-1 replication in acutely infected PBMCs at a step in the viral replicative cycle after reverse transcription, but before or during viral protein expression through a cGMP-independent mechanism. In the latently infected U1 cell line, NO donor compounds and intracellular NO production stimulate HIV-1 reactivation. These studies suggest that NO both inhibits HIV-1 replication in acutely infected cells and stimulates HIV-1 reactivation in chronically infected cells. Thus, NO may have a physiologic role in HIV-1 replication, and NO donor compounds, which have been used for decades in the treatment of coronary artery disease with limited toxicity, might be useful in the treatment of HIV-1 disease by inhibiting acute infection, reactivating latent virus, or both.


Assuntos
HIV-1/fisiologia , Leucócitos Mononucleares/virologia , Mercaptoetanol , Óxido Nítrico/fisiologia , S-Nitrosotióis , Replicação Viral/fisiologia , Adulto , Fármacos Anti-HIV/farmacologia , Linhagem Celular , Células Cultivadas , GMP Cíclico/metabolismo , DNA Viral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/genética , Humanos , Óxido Nítrico/biossíntese , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/biossíntese , Compostos Nitrosos/farmacologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , S-Nitroso-N-Acetilpenicilamina , Transcrição Gênica , Proteínas Virais/biossíntese , Replicação Viral/efeitos dos fármacos , Zidovudina/farmacologia , ômega-N-Metilarginina/farmacologia
18.
J Immunol ; 163(2): 893-7, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10395684

RESUMO

Little is known regarding the mechanism by which T cells control intraabdominal abscess formation. Treating animals with polysaccharide A (PS A) from Bacteroides fragilis shortly before or after challenge protects against abscess formation subsequent to challenge with different abscess-inducing bacteria. Although bacterial polysaccharides are considered to be T cell-independent Ags, T cells from PS A-treated animals mediate this protective activity. In the present study, we demonstrate that CD4+ T cells transfer PS A-mediated protection against abscess formation, and that a soluble mediator produced by these cells confers this activity. Cytokine mRNA analysis showed that T cells from PS A-treated animals produced transcript for IL-2, IFN-gamma, and IL-10, but not for IL-4. The addition of IL-2-specific Ab to T cell lysates taken from PS A-treated animals abrogated the ability to transfer protection, whereas the addition of Abs specific for IFN-gamma and IL-10 did not affect protection. Finally, administration of rIL-2 to animals at the time of bacterial challenge prevented abscess formation in a dose-dependent manner. These data demonstrate that PS A-mediated protection against abscess formation is dependent upon a CD4+ T cell-dependent response, and that IL-2 is essential to this immune mechanism.


Assuntos
Abscesso Abdominal/imunologia , Interleucina-2/fisiologia , Sepse/imunologia , Abscesso Abdominal/prevenção & controle , Transferência Adotiva , Animais , Especificidade de Anticorpos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/genética , Citocinas/imunologia , Soros Imunes/farmacologia , Injeções Intraperitoneais , Interleucina-2/uso terapêutico , Masculino , Polissacarídeos Bacterianos/administração & dosagem , Polissacarídeos Bacterianos/imunologia , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Proteínas Recombinantes/uso terapêutico , Sepse/prevenção & controle , Solubilidade
19.
J Gen Virol ; 80 ( Pt 6): 1519-1527, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10374971

RESUMO

Adenovirus (Ad) tropism is mediated in part through the fibre protein. The common coxsackie B virus and Ad receptor (CAR) was recently identified as the major receptor for subgroup C Ad serotype 5 (Ad5) and serotype 2 (Ad2) fibres. Effects of mutations in the Ad5 fibre gene were studied to assess domains of the fibre capsomer that could alter virus tropism without altering virus assembly and replication. All mutants that accumulated as fibre monomers failed to assemble with a penton base and proved lethal for Ad5 which suggests that the absence of infectious virions resulted in part from a defect in fibre penton base assembly. Cell binding capacity of all fibre mutants was investigated in cell binding competition experiments with adenovirions using CHO-CAR cells (CHO cells that have been transfected with CAR cDNA and express functional CAR). The results suggest that the R-sheet of the Ad5 fibre knob monomer contains binding motifs for CAR and that beta-strands E and F, or a region close to them, may also be involved in receptor recognition.


Assuntos
Adenovírus Humanos/metabolismo , Proteínas do Capsídeo , Capsídeo/genética , Capsídeo/metabolismo , Receptores Virais/metabolismo , Adenovírus Humanos/classificação , Adenovírus Humanos/genética , Animais , Baculoviridae/genética , Ligação Competitiva , Células CHO , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Cricetinae , Células HeLa , Humanos , Luciferases/metabolismo , Mutagênese Sítio-Dirigida , Fenótipo , Plasmídeos/genética , Proteínas Recombinantes/metabolismo , Spodoptera
20.
J Immunol ; 162(10): 6016-23, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10229841

RESUMO

The level of ongoing HIV-1 replication within an individual is critical to HIV-1 pathogenesis. Among host immune factors, the cytokine TNF-alpha has previously been shown to increase HIV-1 replication in various monocyte and T cell model systems. Here, we demonstrate that signaling through the TNF receptor family member, the lymphotoxin-beta (LT-beta) receptor (LT-betaR), also regulates HIV-1 replication. Furthermore, HIV-1 replication is cooperatively stimulated when the distinct LT-betaR and TNF receptor systems are simultaneously engaged by their specific ligands. Moreover, in a physiological coculture cellular assay system, we show that membrane-bound TNF-alpha and LT-alpha1beta2 act virtually identically to their soluble forms in the regulation of HIV-1 replication. Thus, cosignaling via the LT-beta and TNF-alpha receptors is probably involved in the modulation of HIV-1 replication and the subsequent determination of HIV-1 viral burden in monocytes. Intriguingly, surface expression of LT-alpha1beta2 is up-regulated on a T cell line acutely infected with HIV-1, suggesting a positive feedback loop between HIV-1 infection, LT-alpha1beta2 expression, and HIV-1 replication. Given the critical role that LT-alpha1beta2 plays in lymphoid architecture, we speculate that LT-alpha1beta2 may be involved in HIV-associated abnormalities of the lymphoid organs.


Assuntos
HIV-1/crescimento & desenvolvimento , Linfotoxina-alfa/metabolismo , Monócitos/virologia , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Antígenos CD/metabolismo , Linhagem Celular , Sinergismo Farmacológico , Humanos , Receptor beta de Linfotoxina , Linfotoxina-alfa/farmacologia , Proteínas de Membrana/metabolismo , NF-kappa B/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral , Transdução de Sinais , Fator de Necrose Tumoral alfa/farmacologia , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...