Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38871558

RESUMO

BACKGROUND: Talquetamab is a bispecific antibody targeting the multiple myeloma-associated antigen G protein-coupled receptor family C group 5 member D (GPRC5D). In the phase 1/2 MonumenTAL-1 trial (NCT03399799/NCT04634552), overall responses rates were > 71% in patients with triple-class exposed relapsed/refractory multiple myeloma (RRMM). Due to the distribution of the target antigen, a unique pattern of GPRC5D-associated adverse events (AEs) was observed, together with T-cell redirection-associated AEs. Management strategies for talquetamab-associated AEs are described. DISCUSSION: GPRC5D-associated AEs included dermatologic (rash, nonrash, and nail toxicities) and oral AEs (dysgeusia, dysphagia, and dry mouth). The incidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were consistent with other T-cell redirection therapies. The incidence of high-grade infections was lower than observed with B-cell maturation antigen-targeting bispecific antibodies, with less frequent use of intravenous immunoglobulin required. GPRC5D-associated AEs were mostly low grade and led to few discontinuations. Skin toxicities were managed with emollients, topical corticosteroids, and oral corticosteroids (for high-grade, persistent, or AEs that progress). Nail toxicities were commonly managed with emollients. Based on investigator experience, dose modification may be effective for controlling oral events. Observation for potential weight changes is required. Infections were managed per standard of care. CRS and ICANS were effectively managed, consistent with other trials of T-cell redirection therapies. CONCLUSION: Although talquetamab had a distinct safety profile, AEs were considered clinically manageable and mostly low grade. With appropriate education and support, health care practitioners can ensure patients with RRMM maintain quality of life and treatment adherence.

3.
Blood Cancer J ; 14(1): 24, 2024 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-38307865

RESUMO

Multiple myeloma is a genetically complex and heterogenous malignancy with a 5-year survival rate of approximately 60%. Despite advances in therapy, patients experience cycles of remission and relapse, with each successive line of therapy associated with poorer outcomes; therefore, therapies with different mechanisms of action against new myeloma antigens are needed. G protein-coupled receptor class C group 5 member D (GPRC5D) has emerged as a novel therapeutic target for the treatment of multiple myeloma. We review the biology and target validation of GPRC5D, and clinical data from early phase trials of GPRC5D-targeting bispecific antibodies, talquetamab and forimtamig, and chimeric antigen receptor T cell (CAR-T) therapies, MCARH109, OriCAR-017, and BMS-986393. In addition to adverse events (AEs) associated with T-cell-redirection therapies irrespective of target, a consistent pattern of dermatologic and oral AEs has been reported across several trials of GPRC5D-targeting bispecific antibodies, as well as rare cerebellar events with CAR-T therapy. Additional studies are needed to understand the underlying mechanisms involved in the development of skin- and oral-related toxicities. We review the strategies that have been used to manage these GPRC5D-related toxicities. Preliminary efficacy data showed overall response rates for GPRC5D-targeting T-cell-redirecting therapies were ≥64%; most responders achieved a very good partial response or better. Pharmacokinetics/pharmacodynamics showed that these therapies led to cytokine release and T-cell activation. In conclusion, results from early phase trials of GPRC5D-targeting T-cell-redirecting agents have shown promising efficacy and manageable safety profiles, including lower infection rates compared with B-cell maturation antigen- and Fc receptor-like protein 5-targeting bispecific antibodies. Further clinical trials, including those investigating GPRC5D-targeting T-cell-redirecting agents in combination with other anti-myeloma therapies and with different treatment modalities, may help to elucidate the future optimal treatment regimen and sequence for patients with multiple myeloma and improve survival outcomes. Video Summary.


Assuntos
Anticorpos Biespecíficos , Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Humanos , Mieloma Múltiplo/tratamento farmacológico , Anticorpos Biespecíficos/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Imunoterapia Adotiva/métodos , Receptores Acoplados a Proteínas G
4.
JTO Clin Res Rep ; 2(2): 100104, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34589982

RESUMO

INTRODUCTION: The programmed death-ligand 1 inhibitor atezolizumab improves progression-free survival (PFS) and overall survival (OS) for patients with previously treated advanced NSCLC. Preclinical studies indicate that targeting CD38-positive cells with daratumumab may synergistically enhance atezolizumab's antitumor activity by increasing the effector T-cell activity. METHODS: This phase 1b-2 study included a safety run-in (one cycle of daratumumab plus atezolizumab) and randomized phases (daratumumab plus atezolizumab versus atezolizumab alone). The primary objective of the randomized phase was to compare overall response rates. The secondary objectives included evaluations of safety, clinical benefit rate (stable disease or better), PFS, OS, and pharmacokinetics. RESULTS: In total, 99 patients were enrolled (safety run-in, n = 7; randomized, n = 46 per arm). In the randomized phase, the overall response rate was 4.3% for daratumumab plus atezolizumab and 13.0% for atezolizumab alone (OR: 0.30; 95% confidence interval: 0.03-1.92). The respective clinical benefit rates were 52.2% and 43.5%. No improvements were observed in the median PFS or median OS for combination therapy. The study was terminated because of the limited efficacy of daratumumab plus atezolizumab. CONCLUSIONS: Daratumumab plus atezolizumab therapy did not improve efficacy versus atezolizumab monotherapy for patients with previously treated advanced NSCLC.

6.
Protein Eng Des Sel ; 28(10): 385-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26275855

RESUMO

A number of classes of proteins have been engineered for high stability using consensus sequence design methods. Here we describe the engineering of a novel albumin binding domain (ABD) three-helix bundle protein. The resulting engineered ABD molecule, called ABDCon, is expressed at high levels in the soluble fraction of Escherichia coli and is highly stable, with a melting temperature of 81.5°C. ABDCon binds human, monkey and mouse serum albumins with affinity as high as 61 pM. The solution structure of ABDCon is consistent with the three-helix bundle design and epitope mapping studies enabled a precise definition of the albumin binding interface. Fusion of a 10 kDa scaffold protein to ABDCon results in a long terminal half-life of 60 h in mice and 182 h in cynomolgus monkeys. To explore the link between albumin affinity and in vivo exposure, mutations were designed at the albumin binding interface of ABDCon yielding variants that span an 11 000-fold range in affinity. The PK properties of five such variants were determined in mice in order to demonstrate the tunable nature of serum half-life, exposure and clearance with variations in albumin binding affinity.


Assuntos
Albuminas/metabolismo , Sequência Consenso , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Sequência de Aminoácidos , Animais , Escherichia coli/genética , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
7.
J Allergy Clin Immunol ; 132(4): 933-41, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23810766

RESUMO

BACKGROUND: IL-25 and IL-33 belong to distinct cytokine families, but experimental mouse studies suggest their immunologic functions in type 2 immunity are almost entirely overlapping. However, only polymorphisms in the IL-33 pathway (IL1RL1 and IL33) have been significantly associated with asthma in large-cohort genome-wide association studies. OBJECTIVE: We sought to identify distinct pathways for IL-25 and IL-33 in the lung that might provide insight into their roles in asthma pathogenesis and potential for therapeutic intervention. METHODS: IL-25 receptor-deficient (Il17rb(-/-)), IL-33 receptor-deficient (ST2, Il1rl1(-/-)), and double-deficient (Il17rb(-/-)Il1rl1(-/-)) mice were analyzed in models of allergic asthma. Microarrays, an ex vivo lung slice airway contraction model, and Il13(+/eGFP) mice were then used to identify specific effects of IL-25 and IL-33 administration. RESULTS: Comparison of IL-25 and IL-33 pathway-deficient mice demonstrates that IL-33 signaling plays a more important in vivo role in airways hyperreactivity than IL-25. Furthermore, methacholine-induced airway contraction ex vivo increases after treatment with IL-33 but not IL-25. This is dependent on expression of the IL-33 receptor and type 2 cytokines. Confocal studies with Il13(+/eGFP) mice show that IL-33 more potently induces expansion of IL-13-producing type 2 innate lymphoid cells, correlating with airway contraction. This predominance of IL-33 activity is enforced in vivo because IL-33 is more rapidly expressed and released in comparison with IL-25. CONCLUSION: Our data demonstrate that IL-33 plays a critical role in the rapid induction of airway contraction by stimulating the prompt expansion of IL-13-producing type 2 innate lymphoid cells, whereas IL-25-induced responses are slower and less potent.


Assuntos
Asma/fisiopatologia , Hiper-Reatividade Brônquica/imunologia , Interleucina-13/biossíntese , Interleucinas/imunologia , Linfócitos/imunologia , Células Th2/imunologia , Animais , Asma/imunologia , Hiper-Reatividade Brônquica/fisiopatologia , Humanos , Interleucina-33 , Interleucinas/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células Th2/citologia , Células Th2/metabolismo
8.
Inflamm Allergy Drug Targets ; 12(2): 81-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23517644

RESUMO

Inhaled therapeutics are used routinely to treat a variety of pulmonary diseases including asthma, COPD and cystic fibrosis. In addition, biological therapies represent the fastest growing segment of approved pharmaceuticals. However, despite the increased availability of biological therapies, nearly all inhaled therapeutics are small molecule drugs with only a single inhaled protein therapeutic approved. There remains a significant unmet need for therapeutics in pulmonary diseases, and biological therapies with potential to alter disease progression represent a significant opportunity to treat these challenging diseases. This review provides a background into efforts to develop inhaled biological therapies and highlights some of the associated challenges. In addition, we speculate on the ideal properties of a biologic therapy for inhaled delivery.


Assuntos
Produtos Biológicos/administração & dosagem , Pneumopatias/tratamento farmacológico , Proteínas/administração & dosagem , Administração por Inalação , Animais , Humanos
9.
Nature ; 464(7293): 1367-70, 2010 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-20200518

RESUMO

Innate immunity provides the first line of defence against invading pathogens and provides important cues for the development of adaptive immunity. Type-2 immunity-responsible for protective immune responses to helminth parasites and the underlying cause of the pathogenesis of allergic asthma-consists of responses dominated by the cardinal type-2 cytokines interleukin (IL)4, IL5 and IL13 (ref. 5). T cells are an important source of these cytokines in adaptive immune responses, but the innate cell sources remain to be comprehensively determined. Here, through the use of novel Il13-eGFP reporter mice, we present the identification and functional characterization of a new innate type-2 immune effector leukocyte that we have named the nuocyte. Nuocytes expand in vivo in response to the type-2-inducing cytokines IL25 and IL33, and represent the predominant early source of IL13 during helminth infection with Nippostrongylus brasiliensis. In the combined absence of IL25 and IL33 signalling, nuocytes fail to expand, resulting in a severe defect in worm expulsion that is rescued by the adoptive transfer of in vitro cultured wild-type, but not IL13-deficient, nuocytes. Thus, nuocytes represent a critically important innate effector cell in type-2 immunity.


Assuntos
Imunidade Inata/imunologia , Interleucinas/imunologia , Leucócitos/imunologia , Células Th2/imunologia , Transferência Adotiva , Animais , Células Cultivadas , Interleucina-13/biossíntese , Interleucina-13/deficiência , Interleucina-13/genética , Interleucina-17/deficiência , Interleucina-17/genética , Interleucinas/biossíntese , Interleucinas/deficiência , Interleucinas/genética , Leucócitos/citologia , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nippostrongylus/imunologia , Infecções por Strongylida/imunologia
10.
Immunology ; 126(1): 28-34, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19120496

RESUMO

Dendritic cells (DCs) play a central role in activating CD4 T (T helper, Th) cells. As a component of their response to pathogen-associated stimuli, DCs produce cytokines and express surface molecules that provide important cues to modulate the effector functions of responding Th cells. Much is known of how DCs respond to, and influence immune response outcome to, bacterial and viral pathogens. However, relatively little is understood about how DCs respond to helminth parasites. This is an area of considerable interest since it impacts our understanding of the initiation of Th2 responses, which are stereotypically associated with helminth infections, and the regulation of allergic and autoimmune pathologies which evidence suggests are less severe or absent in individuals infected with helminths. This review attempts to summarize our understanding of the effects of helminth products on dendritic cell biology.


Assuntos
Células Dendríticas/imunologia , Helmintos/imunologia , Animais , Antígenos de Helmintos/imunologia , Helmintíase/imunologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Higiene , Camundongos , Células Th2/imunologia , Receptores Toll-Like/imunologia
11.
Infect Immun ; 76(12): 5754-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18824534

RESUMO

Unlike most pathogens, helminth parasites and their products induce strong Th2 responses, and dendritic cells (DCs) and macrophages exposed to helminth antigens generally fail to produce interleukin-12. Rather, it has been shown that helminth products such as soluble egg antigens (SEA; a soluble extract from Schistosoma mansoni eggs) inhibit the activation of DCs in response to classical Toll-like receptor (TLR) ligands such as lipopolysaccharide or CpG. Nevertheless, recent work has suggested that TLR4 and/or TLR2 plays an important role in the recognition of helminth products by DCs and macrophages and in the development of Th2 responses. Using DCs derived from TLR4(-/-), TLR2(-/-), or MyD88(-/-) mice, we have demonstrated that the ability of SEA to modulate DC activation is MyD88 independent and requires neither TLR4 nor TLR2. Moreover, TLR2 and TLR4 are not required for SEA-pulsed DCs to induce Th2 responses in naïve mice.


Assuntos
Antígenos de Helmintos/imunologia , Proteínas de Helminto/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Esquistossomose mansoni/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Citocinas/biossíntese , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Schistosoma mansoni/imunologia , Células Th2/imunologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
12.
Chem Immunol Allergy ; 90: 82-90, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16210904

RESUMO

There is increasing awareness that dendritic cells (DCs) can interpret pathogen-inherent signals and play a pivotal role in polarizing Th cell differentiation. Polarized Th1 responses are induced by DCs, which respond to pathogen-derived TLR ligands to mature and produce IL-12 and related cytokines that are instrumental in Th1 cell outgrowth. In contrast, DCs exposed to SEA (soluble egg Ag from the helminth parasite Schistosoma mansoni) retain a (modified) immature phenotype and induce Th2 responses. In addition to providing positive signals for Th1 cell development, DCs activated to mature by TLR-engagement also provide a potent negative signal that prevents the development of Th2 cells. Production of this signal is dependent upon a MyD88-dependent signaling pathway in DCs. In contrast, exposure of DCs to SEA severely limits their ability to respond to inflammatory TLR ligands such as LPS and CpG. Thus as part of their pathogen-specific response programs, DC can exert negative as well as positive signals for Th response polarization. These effects may have powerful and systemic effects on disease outcome.


Assuntos
Células Dendríticas/imunologia , Schistosoma mansoni/química , Schistosoma mansoni/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Diferenciação Celular , Células Dendríticas/citologia , Ativação Linfocitária , Linfócitos T Auxiliares-Indutores/citologia
13.
J Immunol ; 174(11): 7154-9, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15905559

RESUMO

A number of receptors and signaling pathways can influence the ability of dendritic cells (DC) to promote CD4(+) Th type 1 (Th1) responses. In contrast, the regulatory pathways and signaling events that govern the ability of DC to instruct Th2 cell differentiation remain poorly defined. In this report, we demonstrate that NF-kappaB1 expression within DC is required to promote optimal Th2 responses following exposure to Schistosoma mansoni eggs, a potent and natural Th2-inducing stimulus. Although injection of S. mansoni eggs induced production of IL-4, IL-5, and IL-13 in the draining lymph node of wild-type (WT) mice, NF-kappaB1(-/-) hosts failed to express Th2 cytokines and developed a polarized Ag-specific IFN-gamma response. In an in vivo adoptive transfer model in which NF-kappaB-sufficient OVA-specific DO11.10 TCR transgenic T cells were injected into OVA-immunized WT or NF-kappaB1(-/-) hosts, NF-kappaB1(-/-) APCs efficiently promoted CD4(+) T cell proliferation and IFN-gamma responses, but failed to promote Ag-specific IL-4 production. Further, bone marrow-derived DC from NF-kappaB1(-/-) mice failed to promote OVA-specific Th2 cell differentiation in in vitro coculture studies. Last, S. mansoni egg Ag-pulsed NF-kappaB1(-/-) DC failed to prime for Th2 cytokine responses following injection into syngeneic WT hosts. Impaired Th2 priming by NF-kappaB1(-/-) DC was accompanied by a reduction in MAPK phosphorylation in Ag-pulsed DC. Taken together, these studies identify a novel requirement for DC-intrinsic expression of NF-kappaB1 in regulating the MAPK pathway and governing the competence of DC to instruct Th2 cell differentiation.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , NF-kappa B/biossíntese , Precursores de Proteínas/biossíntese , Células Th2/imunologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/fisiologia , Animais , Antígenos de Helmintos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Células Dendríticas/patologia , Epitopos de Linfócito T/imunologia , Proteínas I-kappa B/biossíntese , Proteínas I-kappa B/genética , Proteínas I-kappa B/fisiologia , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/deficiência , NF-kappa B/genética , NF-kappa B/fisiologia , Subunidade p50 de NF-kappa B , Fosforilação , Precursores de Proteínas/deficiência , Precursores de Proteínas/genética , Precursores de Proteínas/fisiologia , Schistosoma mansoni/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Th2/metabolismo , Células Th2/parasitologia , Células Th2/patologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/deficiência
14.
J Immunol ; 173(12): 7454-61, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15585871

RESUMO

There is increasing awareness that helminth infections can ameliorate proinflammatory conditions. In part, this is due to their inherent ability to induce Th2 and, perhaps, regulatory T cell responses. However, recent evidence indicates that helminths also have direct anti-inflammatory effects on innate immune responses. In this study, we address this issue and show that soluble molecules from the eggs of the helminth parasite Schistosoma mansoni (SEA) suppress LPS-induced activation of immature murine dendritic cells, including MHC class II, costimulatory molecule expression, and IL-12 production. SEA-augmented LPS-induced production of IL-10 is in part responsible for the observed reduction in LPS-induced IL-12 production. However, analyses of IL-10(-/-) DC revealed distinct IL-10-independent suppressive effects of SEA. IL-10-independent mechanisms are evident in the suppression of TLR ligand-induced MAPK and NF-kappaB signaling pathways. Microarray analyses demonstrate that SEA alone uniquely alters the expression of a small subset of genes that are not up-regulated during conventional TLR-induced DC maturation. In contrast, the effects of SEA on TLR ligand-induced DC activation were striking: when mixed with LPS, SEA significantly affects the expression of >100 LPS-regulated genes. These findings indicate that SEA exerts potent anti-inflammatory effects by directly regulating the ability of DC to respond to TLR ligands.


Assuntos
Antígenos de Helmintos/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Fatores Imunológicos/fisiologia , Glicoproteínas de Membrana/fisiologia , Receptores de Superfície Celular/fisiologia , Schistosoma mansoni/imunologia , Animais , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/parasitologia , Regulação para Baixo/imunologia , Regulação da Expressão Gênica/imunologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Interleucina-10/biossíntese , Interleucina-10/fisiologia , Interleucina-12/antagonistas & inibidores , Interleucina-12/biossíntese , Ligantes , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Superfície Celular/antagonistas & inibidores , Transdução de Sinais/imunologia , Receptores Toll-Like , Regulação para Cima/imunologia
15.
Immunol Rev ; 201: 117-26, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15361236

RESUMO

T-helper 2 (Th2) cell responses play a critical role in protection against helminth infections. In the case of Schistosoma mansoni, an important helminth parasite of man, data from a mouse model of human disease have shown that Th2 responses are essential to allow host survival. In this infection, parasite eggs are the primary stimulus for Th2 response development. Recent work has shown that egg molecules exert multiple levels of control over the development of host interferon-gamma-associated inflammatory responses. Soluble egg antigen inhibits the ability of dendritic cells to make interleukin-12 and induces Th2-polarized adaptive immune responses that in combination with regulatory T-cell responses effectively limit Th1 response development. In this article, we discuss the factors influencing Th2 response polarization during infection with S. mansoni.


Assuntos
Schistosoma mansoni/imunologia , Células Th2/imunologia , Animais , Antígenos de Helmintos/imunologia , Humanos , Camundongos , Schistosoma mansoni/crescimento & desenvolvimento , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia
16.
J Immunol ; 172(4): 2016-20, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14764665

RESUMO

To examine the ability of dendritic cells (DC) to discriminate between helminth and microbial Ag and induce appropriately polarized Th responses, mouse DC were copulsed with the helminth Ag, schistosome egg Ag (SEA), along with the bacterium Proprionebacterium acnes, Pa, and transferred into wild-type mice. Strikingly, SEA/Pa-copulsed DC induced concurrent Pa-specific Th1 (but not Th2) responses and SEA-specific Th2 (but not Th1) responses. Although DC exposed to both Ag undergo many of the maturation-associated changes that accompany exposure to Pa alone, Pa-induced IL-12 production was inhibited by SEA. Examination of Ag uptake revealed that SEA and Pa are acquired via discrete pathways and enter nonoverlapping intracellular compartments. Data suggest that segregation of SEA and Pa into distinct compartments, coupled with SEA-induced modifications of the DC maturation pathway, are significant components of the ability of DC to interpret signals inherent to SEA and Pa and induce appropriately polarized Th responses.


Assuntos
Antígenos de Bactérias/imunologia , Antígenos de Helmintos/imunologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Líquido Intracelular/imunologia , Schistosoma/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Apresentação de Antígeno , Antígenos de Bactérias/metabolismo , Antígenos de Helmintos/metabolismo , Antígeno B7-1/biossíntese , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/microbiologia , Células Dendríticas/parasitologia , Células Dendríticas/transplante , Feminino , Antígenos de Histocompatibilidade Classe II/biossíntese , Líquido Intracelular/microbiologia , Líquido Intracelular/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Propionibacterium acnes/imunologia , Células Th1/microbiologia , Células Th1/parasitologia , Células Th2/microbiologia , Células Th2/parasitologia
17.
J Bacteriol ; 184(16): 4409-19, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12142411

RESUMO

The mxi-spa locus on the virulence plasmid of Shigella flexneri encodes components of the type III secretion system. mxiE, a gene within this locus, encodes a protein that is homologous to the AraC/XylS family of transcriptional regulators, but currently its role in pathogenesis remains undefined. We characterized the virulence phenotype of a nonpolar mxiE mutant and found that this mutant retained the ability to invade mammalian cells in tissue culture and secrete Ipas (type III effectors required for host cell invasion), although it was less efficient than wild-type Shigella at cell-to-cell spread. Despite its invasive properties in culture, the mxiE mutant was completely avirulent in an animal model. Potential targets for MxiE activation were identified by using promoter-green fluorescent protein fusions, and gene expression was examined under various growth conditions. Six MxiE-regulated genes were discovered: ospB, ospC1, ospE2, ospF, virA, and ipaH(9.8). Notably, activation of these genes only occurred within the intracellular environment of the host and not during growth at 37 degrees C in liquid culture. Interestingly, all of the MxiE-regulated proteins previously have been shown to be secreted through the type III secretion system and are putative virulence factors. Our findings suggest that some of these Osp proteins may be involved in postinvasion events related to virulence. Since bacterial pathogens adapt to multiple environments during the course of infecting a host, we propose that Shigella evolved a mechanism to take advantage of a unique intracellular cue, which is mediated through MxiE, to express proteins when the organism reaches the eukaryotic cytosol.


Assuntos
Antígenos de Bactérias , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Ligação a DNA , Lipoproteínas , Shigella flexneri/metabolismo , Shigella flexneri/patogenicidade , Fatores de Transcrição , Fatores de Virulência , Proteínas de Bactérias/genética , Genes araC/genética , Mutagênese/fisiologia , Fenótipo , Plasmídeos/genética , Shigella flexneri/genética , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA