Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 25(5): 873-885, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38553615

RESUMO

Metabolic programming is important for B cell fate, but the bioenergetic requirement for regulatory B (Breg) cell differentiation and function is unknown. Here we show that Breg cell differentiation, unlike non-Breg cells, relies on mitochondrial electron transport and homeostatic levels of reactive oxygen species (ROS). Single-cell RNA sequencing analysis revealed that TXN, encoding the metabolic redox protein thioredoxin (Trx), is highly expressed by Breg cells, unlike Trx inhibitor TXNIP which was downregulated. Pharmacological inhibition or gene silencing of TXN resulted in mitochondrial membrane depolarization and increased ROS levels, selectively suppressing Breg cell differentiation and function while favoring pro-inflammatory B cell differentiation. Patients with systemic lupus erythematosus (SLE), characterized by Breg cell deficiencies, present with B cell mitochondrial membrane depolarization, elevated ROS and fewer Trx+ B cells. Exogenous Trx stimulation restored Breg cells and mitochondrial membrane polarization in SLE B cells to healthy B cell levels, indicating Trx insufficiency underlies Breg cell impairment in patients with SLE.


Assuntos
Proteínas de Transporte , Diferenciação Celular , Lúpus Eritematoso Sistêmico , Mitocôndrias , Espécies Reativas de Oxigênio , Tiorredoxinas , Tiorredoxinas/metabolismo , Tiorredoxinas/genética , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Mitocôndrias/metabolismo , Feminino , Animais , Camundongos , Potencial da Membrana Mitocondrial , Masculino , Adulto , Oxirredução
2.
J Allergy Clin Immunol ; 152(5): 1237-1246, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37536512

RESUMO

BACKGROUND: Few studies have explored the immunology and genetic risk of paradoxical eczema occurring as an adverse event of biologic therapy in patients with psoriasis. OBJECTIVES: We sought to describe the systemic inflammatory signature of paradoxical eczema using proteomics and explore whether this is genetically mediated. METHODS: This study used the Olink Target 96 Inflammation panel on 256 serum samples from 71 patients with psoriasis and paradoxical eczema, and 75 controls with psoriasis to identify differentially expressed proteins and enriched gene sets. Case samples from 1 or more time points (T1 prebiologic, T2 postbiologic, and T3 postparadoxical eczema) were matched 1:1 with control samples. Genes contributing to enriched gene sets were selected, and functional single nucleotide polymorphisms used to create polygenic risk scores in a genotyped cohort of 88 paradoxical eczema cases and 3124 psoriasis controls. RESULTS: STAMBP expression was lower in cases at T1 than in controls (log-fold change: -0.44; adjusted P = .022); no other proteins reached statistical significance at equivalent time points. Eleven gene sets including cytokine and chemokine pathways were enriched in cases at T2 and 10 at T3. Of the 39 proteins contributing to enriched gene sets, the majority are associated with the atopic dermatitis serum proteome. A polygenic risk score including 38 functional single nucleotide polymorphisms linked to enriched gene sets was associated with paradoxical eczema (adjusted P = .046). CONCLUSIONS: The paradoxical eczema systemic inflammatory proteome trends toward atopic dermatitis at a gene-set level and is detectable before onset of the phenotype. This signature could be genetically determined.


Assuntos
Produtos Biológicos , Dermatite Atópica , Eczema , Psoríase , Humanos , Dermatite Atópica/genética , Proteômica , Proteoma , Psoríase/tratamento farmacológico , Psoríase/genética , Genômica , Eczema/genética
3.
Eur J Immunol ; 45(6): 1842-54, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25763771

RESUMO

B cells have been described as having the capacity to regulate cellular immune responses and suppress inflammatory processes. One such regulatory B-cell population is defined as IL-10-producing CD19(+) CD1d(hi) cells. Previous work has identified an expansion of these cells in mice infected with the helminth, Schistosoma mansoni. Here, microarray analysis of CD19(+) CD1d(hi) B cells from mice infected with S. mansoni demonstrated significantly increased Tlr7 expression, while CD19(+) CD1d(hi) B cells from uninfected mice also demonstrated elevated Tlr7 expression. Using IL-10 reporter, Il10(-/-) and Tlr7(-/-) mice, we formally demonstrate that TLR7 ligation of CD19(+) CD1d(hi) B cells increases their capacity to produce IL-10. In a mouse model of allergic lung inflammation, the adoptive transfer of TLR7-elicited CD19(+) CD1d(hi) B cells reduced airway inflammation and associated airway hyperresponsiveness. Using DEREG mice to deplete FoxP3(+) T regulatory cells in allergen-sensitized mice, we show that that TLR7-elicited CD19(+) CD1d(hi) B cells suppress airway hyperresponsiveness via a T regulatory cell dependent mechanism. These studies identify that TLR7 stimulation leads to the expansion of IL-10-producing CD19(+) CD1d(hi) B cells, which can suppress allergic lung inflammation via T regulatory cells.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Pneumonia/imunologia , Pneumonia/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Receptor 7 Toll-Like/metabolismo , Animais , Antígenos CD19/metabolismo , Antígenos CD1d/metabolismo , Modelos Animais de Doenças , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Interleucina-10/biossíntese , Camundongos , Camundongos Knockout , Ovalbumina/efeitos adversos , Pneumonia/parasitologia , Ligação Proteica , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/parasitologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/metabolismo , Regulação para Cima
4.
JAMA Dermatol ; 160(1): 71-79, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38055239

RESUMO

Importance: Biologics used for plaque psoriasis have been reported to be associated with an atopic dermatitis (AD) phenotype, or paradoxical eczema, in some patients. The risk factors for this are unknown. Objective: To explore risk of paradoxical eczema by biologic class and identify factors associated with paradoxical eczema. Design, Setting, and Participants: This prospective cohort study used data from the British Association of Dermatologists Biologics and Immunomodulators Register for adults treated with biologics for plaque psoriasis who were seen at multicenter dermatology clinics in the UK and Ireland. Included participants were registered and had 1 or more follow-up visits between September 2007 and December 2022. Exposures: Duration of exposure to tumor necrosis factor (TNF) inhibitors, interleukin (IL) 17 inhibitors, IL-12/23 inhibitors, or IL-23 inhibitors until paradoxical eczema onset, treatment discontinuation, last follow-up, or death. Main Outcomes and Measures: Incidence rates of paradoxical eczema, paradoxical eczema risk by biologic class, and the association of demographic and clinical variables with risk of paradoxical eczema were assessed using propensity score-weighted Cox proportional hazards regression models. Results: Of 56 553 drug exposures considered, 24 997 from 13 699 participants were included. The 24 997 included exposures (median age, 46 years [IQR, 36-55 years]; 57% male) accrued a total exposure time of 81 441 patient-years. A total of 273 exposures (1%) were associated with paradoxical eczema. The adjusted incidence rates were 1.22 per 100 000 person-years for IL-17 inhibitors, 0.94 per 100 000 person-years for TNF inhibitors, 0.80 per 100 000 person-years for IL-12/23 inhibitors, and 0.56 per 100 000 person-years for IL-23 inhibitors. Compared with TNF inhibitors, IL-23 inhibitors were associated with a lower risk of paradoxical eczema (hazard ratio [HR], 0.39; 95% CI, 0.19-0.81), and there was no association of IL-17 inhibitors (HR, 1.03; 95% CI, 0.74-1.42) or IL-12/23 inhibitors (HR, 0.87; 95% CI, 0.66-1.16) with risk of paradoxical eczema. Increasing age (HR, 1.02 per year; 95% CI, 1.01-1.03) and history of AD (HR, 12.40; 95% CI, 6.97-22.06) or hay fever (HR, 3.78; 95% CI, 1.49-9.53) were associated with higher risk of paradoxical eczema. There was a lower risk in males (HR, 0.60; 95% CI, 0.45-0.78). Conclusions and Relevance: In this study, in biologic-treated patients with psoriasis, paradoxical eczema risk was lowest in patients receiving IL-23 inhibitors. Increasing age, female sex, and history of AD or hay fever were associated with higher risk of paradoxical eczema. The overall incidence of paradoxical eczema was low. Further study is needed to replicate these findings.


Assuntos
Produtos Biológicos , Eczema , Psoríase , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores Biológicos/efeitos adversos , Produtos Biológicos/efeitos adversos , Dermatite Atópica , Eczema/induzido quimicamente , Eczema/epidemiologia , Interleucina-12 , Interleucina-17 , Interleucina-23 , Estudos Prospectivos , Psoríase/tratamento farmacológico , Psoríase/epidemiologia , Rinite Alérgica Sazonal , Inibidores do Fator de Necrose Tumoral/uso terapêutico
5.
Mol Cancer ; 12: 23, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23531080

RESUMO

BACKGROUND: Neuroblastoma is responsible for 15% of all childhood cancer deaths. Despite advances in treatment and disease management, the overall 5-year survival rates remain poor in high-risk disease (25-40%). MiR-497 was previously identified by our laboratory as a member of a miRNA expression signature, predictive of neuroblastoma patient survival and has been reported as a tumor suppressor in a variety of other cancers. WEE1, a tyrosine kinase regulator of the cell cycle and predicted target of miR-497, has emerged as an oncogene in several cancer types and therefore represents an attractive potential target for novel therapy approaches in high-risk neuroblastoma. Our aim was to investigate the potential tumor suppressive role of miR-497 in high-risk neuroblastoma. METHODS: Expression levels of miR-497 and WEE1 in tissues and cells were determined using RT-PCR. The effect of miR-497 and siWEE1 on cell viability was evaluated using MTS assays, apoptosis levels were determined using FACS analysis of Annexin V/PI stained cells, and target protein expression was determined using western blot. Luciferase reporter plasmids were constructed to confirm direct targeting. Results were reported as mean±S.E.M and differences were tested for significance using 2-tailed Students t-test. RESULTS: We determined that miR-497 expression was significantly lower in high-risk MYCN amplified (MNA) tumors and that low miR-497 expression was associated with worse EFS and OS in our cohort. Over-expression of miR-497 reduced cell viability and increased apoptosis in MNA cells. We identified WEE1 as a novel target for miR-497 in neuroblastoma. Furthermore, our analysis showed that high WEE1 levels are significantly associated with poor EFS and OS in neuroblastoma and that siRNA knockdown of WEE1 in MNA cell lines results in significant levels of apoptosis, supporting an oncogenic role of WEE1 in neuroblastoma. Cisplatin (CDDP) treatment of both miR-497 over-expressing cells and WEE1 inhibited cells, resulted in a significant increase in apoptosis in MNA cells, describing a synergistic effect and therefore a potential therapeutic for high-risk neuroblastoma. CONCLUSION: Our study's results are consistent with miR-497 being a candidate tumor suppressor in neuroblastoma, through the direct targeting of WEE1. These findings re-enforce the proposal of WEE1 as a therapeutic target in neuroblastoma.


Assuntos
Proteínas de Ciclo Celular/genética , Amplificação de Genes , MicroRNAs/metabolismo , Neuroblastoma/metabolismo , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Proteínas Tirosina Quinases/genética , Regiões 3' não Traduzidas , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/farmacologia , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Lactente , Estimativa de Kaplan-Meier , MicroRNAs/genética , MicroRNAs/fisiologia , Análise Multivariada , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/mortalidade , Proteínas Nucleares/metabolismo , Modelos de Riscos Proporcionais , Proteínas Tirosina Quinases/metabolismo , RNA Interferente Pequeno/genética
6.
J Invest Dermatol ; 143(8): 1470-1478.e1, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36804406

RESUMO

Biologic therapies for psoriasis can cause paradoxical eczema. The role of genetic factors in its pathogenesis is unknown. To identify risk variants, we conducted a GWAS of 3,212 patients with psoriasis, of whom 88 developed paradoxical eczema. Two lead SNPs reached genome-wide significance (P ≤ 5 × 10-8) for association with paradoxical eczema: rs192705221 (near UNC5B, P = 9.52 × 10-10) and rs72925168 (within SLC1A2, P = 1.66 × 10-9). Genome-wide significant SNPs from published GWAS were used to generate polygenic risk scores (PRSs) for atopic eczema, general atopic disease, or a combination, which were tested for association with paradoxical eczema. Improvement over a clinical risk model was assessed by the area under the curve. All three atopy polygenic risk scores were associated with paradoxical eczema (P < 0.05); polygenic risk score for a combination of atopic eczema and general atopic disease had the strongest association (OR = 1.83, 95% CI = 1.17-2.84, P = 0.0078). Including atopic polygenic risk scores in the multivariable model, which included age, sex, atopic background, and psoriatic arthritis history, increased the area under the curve from 0.671 to 0.681-0.686. Atopic genetic burden is associated with paradoxical eczema occurring in biologic-treated patients with psoriasis, indicating shared underlying mechanisms. Incorporating genetic risk may improve treatment outcome prediction models for psoriasis.


Assuntos
Produtos Biológicos , Dermatite Atópica , Eczema , Psoríase , Humanos , Dermatite Atópica/complicações , Eczema/epidemiologia , Eczema/genética , Psoríase/tratamento farmacológico , Psoríase/genética , Psoríase/complicações , Fatores de Risco , Receptores de Netrina
7.
Clin Transl Gastroenterol ; 12(5): e00360, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34029233

RESUMO

INTRODUCTION: Effective colorectal cancer (CRC) prevention and screening requires sensitive detection of all advanced neoplasias (CRC and advanced adenomas [AA]). However, existing noninvasive screening approaches cannot accurately detect adenomas with high sensitivity. METHODS: Here, we describe a multifactor assay (RNA-FIT test) that combines 8 stool-derived eukaryotic RNA biomarkers, patient demographic information (smoking status), and a fecal immunochemical test (FIT) to sensitively detect advanced colorectal neoplasias and other non-advanced adenomas in a 1,305-patient, average-risk, prospective cohort. This cohort was supplemented with a 22-patient retrospective cohort consisting of stool samples obtained from patients diagnosed with AA or CRC before treatment or resection. Participants within these cohorts were evaluated with the RNA-FIT assay and an optical colonoscopy. RNA-FIT test results were compared with colonoscopy findings. RESULTS: Model performance was assessed through 5-fold internal cross-validation of the training set (n = 939) and by using the model on a hold out testing set (n = 388). When used on the hold out testing set, the RNA-FIT test attained a 95% sensitivity for CRC (n = 22), 62% sensitivity for AA (n = 52), 25% sensitivity for other non-AA (n = 139), 80% specificity for hyperplastic polyps (n = 74), and 85% specificity for no findings on a colonoscopy (n = 101). DISCUSSION: The RNA-FIT assay demonstrated clinically relevant detection of all grades of colorectal neoplasia, including carcinomas, AAs, and ONAs. This assay could represent a noninvasive option to screen for both CRC and precancerous adenomas.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias Colorretais/diagnóstico , Fezes/química , Imunoquímica/métodos , RNA/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Detecção Precoce de Câncer/métodos , Feminino , Humanos , Masculino , Programas de Rastreamento/métodos , Pessoa de Meia-Idade , Estudos Prospectivos , Estudos Retrospectivos , Sensibilidade e Especificidade , Fumar , Fatores Socioeconômicos
8.
J Med Chem ; 61(15): 6705-6723, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-29952567

RESUMO

The primary target of a novel series of immunosuppressive 7-piperazin-1-ylthiazolo[5,4- d]pyrimidin-5-amines was identified as the lipid kinase, PI4KIIIß. Evaluation of the series highlighted their poor solubility and unwanted off-target activities. A medicinal chemistry strategy was put in place to optimize physicochemical properties within the series, while maintaining potency and improving selectivity over other lipid kinases. Compound 22 was initially identified and profiled in vivo, before further modifications led to the discovery of 44 (UCB9608), a vastly more soluble, selective compound with improved metabolic stability and excellent pharmacokinetic profile. A co-crystal structure of 44 with PI4KIIIß was solved, confirming the binding mode of this class of inhibitor. The much-improved in vivo profile of 44 positions it as an ideal tool compound to further establish the link between PI4KIIIß inhibition and prolonged allogeneic organ engraftment, and suppression of immune responses in vivo.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/farmacocinética , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Piperazinas/farmacologia , Piperazinas/farmacocinética , Piperidinas/farmacologia , Transplante Homólogo , Administração Oral , Animais , Disponibilidade Biológica , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/metabolismo , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/metabolismo , Imunossupressores/farmacocinética , Imunossupressores/farmacologia , Camundongos , Simulação de Acoplamento Molecular , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Piperazinas/administração & dosagem , Piperazinas/metabolismo , Piperidinas/administração & dosagem , Piperidinas/metabolismo , Conformação Proteica
9.
J Exp Med ; 214(9): 2507-2521, 2017 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-28747424

RESUMO

Group 2 innate lymphoid cells (ILC2s) are important effector cells driving the initiation of type 2 immune responses leading to adaptive T helper 2 (Th2) immunity. Here we show that ILC2s dynamically express the checkpoint inhibitor molecule PD-L1 during type 2 pulmonary responses. Surprisingly, PD-L1:PD-1 interaction between ILC2s and CD4+ T cells did not inhibit the T cell response, but PD-L1-expressing ILC2s stimulated increased expression of GATA3 and production of IL-13 by Th2 cells both in vitro and in vivo. Conditional deletion of PD-L1 on ILC2s impaired early Th2 polarization and cytokine production, leading to delayed worm expulsion during infection with the gastrointestinal helminth Nippostrongylus brasiliensis Our results identify a novel PD-L1-controlled mechanism for type 2 polarization, with ILC2s mediating an innate checkpoint to control adaptive T helper responses, which has important implications for the treatment of type 2 inflammation.


Assuntos
Antígeno B7-H1/fisiologia , Linfócitos/fisiologia , Células Th2/fisiologia , Imunidade Adaptativa/imunologia , Imunidade Adaptativa/fisiologia , Animais , Antígeno B7-H1/imunologia , Fator de Transcrição GATA3/fisiologia , Imunidade Celular/imunologia , Imunidade Celular/fisiologia , Interleucina-13/fisiologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Nippostrongylus/imunologia , Infecções por Strongylida/imunologia , Células Th2/imunologia
10.
PLoS Negl Trop Dis ; 11(10): e0006012, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29073139

RESUMO

The helminth Schistosoma mansoni modulates the infected host's immune system to facilitate its own survival, by producing excretory/secretory molecules that interact with a variety of the host's cell types including those of the immune system. Herein, we characterise the S. mansoni adult male worm secretome and identify 111 proteins, including 7 vaccine candidates and several molecules with potential immunomodulatory activity. Amongst the molecules present in the secretome, a 17-19kDa protein analogous to human cyclophilin A was identified. Given the ability of cyclophilin A to modulate the immune system by regulating antigen presenting cell activity, we sought to determine whether recombinant S. mansoni Cyclophilin A (rSmCypA) is capable of modulating bone-marrow derived dendritic cell (BMDC) and T cell responses under in vitro conditions. rSmCypA was enzymatically active and able to alter the pro-inflammatory cytokine profile of LPS-activated dendritic cells. rSmCypA also modulated DC function in the induction of CD4+ T cell proliferation with a preferential expansion of Treg cells. This work demonstrates the unique protein composition of the S. mansoni male worm secretome and immunomodulatory activity of S. mansoni Cyclophilin A.


Assuntos
Ciclofilina A/imunologia , Proteínas de Helminto/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/parasitologia , Animais , Ciclofilina A/genética , Células Dendríticas/imunologia , Feminino , Proteínas de Helminto/genética , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , Schistosoma mansoni/genética , Esquistossomose mansoni/imunologia , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia
11.
Nat Commun ; 6: 5997, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25609381

RESUMO

Specific B-cell subsets can regulate T-cell immune responses, and are termed regulatory B cells (Breg). The majority of Breg cells described in mouse and man have been identified by IL-10 production and are known to suppress allergy and autoimmunity. However, Breg cell mediated immune suppression, independent of IL-10, also occurs. Here we show that Breg cells play a critical role in regulating humoral immunity mediated by CD4(+)CXCR5(+)PD-1(+) follicular helper T cells, and can suppress inflammation in autoimmune disease through elevated expression of PD-L1. We have also identified that these B cells are resistant to αCD20 B-cell depletion. This work describes how Breg cells are critical in humoral homoeostasis and may have implications for the regulation of autoimmune diseases.


Assuntos
Linfócitos B Reguladores/citologia , Antígeno B7-H1/biossíntese , Imunidade Humoral/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Adulto , Animais , Anticorpos Monoclonais/química , Autoimunidade , Linfócitos B Reguladores/imunologia , Antígeno B7-H1/fisiologia , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Separação Celular , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Feminino , Homeostase , Humanos , Sistema Imunitário , Imunoglobulina G/imunologia , Inflamação , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR5/metabolismo , Adulto Jovem
12.
Methods Mol Biol ; 1190: 143-62, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25015279

RESUMO

The activation of B cells by pathogens to elicit antibody production is a central facet of immunity. Pathogens also evoke the expansion of B cells that can function in the regulation of immunity. Different pathogens have been described with the capacity to drive such regulatory B (Breg) cells. One group of pathogens, parasitic helminths, has been used experimentally to identify and explore immunological mechanisms of Breg cells. Several species of helminths have demonstrated the capacity to expand Breg cell populations in mice, while the presence of Breg cells in humans infected with certain helminths has also been identified. Herein, we outline how the helminth Schistosoma mansoni can expand Breg cellular responses in vitro. We describe the establishment of a laboratory-based S. mansoni life cycle and methodology for detecting Breg cells via flow cytometry.


Assuntos
Linfócitos B Reguladores/imunologia , Linfócitos B Reguladores/parasitologia , Interações Hospedeiro-Parasita , Schistosoma mansoni/fisiologia , Esquistossomose mansoni/imunologia , Esquistossomose mansoni/parasitologia , Animais , Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Feminino , Citometria de Fluxo/métodos , Técnicas de Genotipagem/métodos , Humanos , Interleucina-10/análise , Interleucina-10/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Schistosoma mansoni/genética , Schistosoma mansoni/crescimento & desenvolvimento , Schistosoma mansoni/imunologia , Caramujos/parasitologia , Baço/citologia , Baço/imunologia , Baço/parasitologia
13.
Int J Parasitol ; 43(3-4): 293-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23291459

RESUMO

The use of live helminth infections is currently in clinical trials as a novel approach for the treatment of a range of allergic and autoimmune diseases. This rapid progression from observational studies some 20 years ago to helminth clinical trials can be attributed to huge advances in not just pre-clinical and clinical evidence, pertaining to the efficacy of these parasites in unrelated diseases, but also a greater understanding of the complex immunological mechanisms that underpin these effects. Helminths have exerted significant evolutionary selective pressures on the host immune genome or "immunome". Studies on helminths were pivotal in a paradigm shift in immunology with recent discoveries of a number of novel immune cell populations. Critically, these new discoveries highlight the need to further understand the underlying mechanism behind the desirable therapeutic effects that helminths offer. With these unknown unknowns there is the distinct possibility that a true, fundamental modus operandi for helminth therapy will arrive long after it has been established in the clinic.


Assuntos
Helmintos/imunologia , Doenças do Sistema Imunitário/terapia , Terapia com Helmintos , Animais , Helmintos/fisiologia , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/parasitologia , Pesquisa Translacional Biomédica
14.
Int Rev Immunol ; 29(5): 461-84, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20839911

RESUMO

Several studies have linked tumor-infiltration by regulatory T cells with poor patient outcome. Targeting the mechanisms by which regulatory T cells traffic to and persist in the tumor may circumvent tumor immune-escape by de-restricting T cell-mediated cytotoxicity. In this review, we describe the principle axes that govern regulatory T cell migration and the mechanisms that underpin their immunosuppressive activity in cancer. Inhibiting either the migration or function of regulatory T cells may enhance host-anti-cancer immune responses and as such are attractive and tractable targets for therapeutic intervention.


Assuntos
Antineoplásicos/uso terapêutico , Imunoterapia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Antineoplásicos/farmacologia , Ensaios Clínicos como Assunto , Citocinas/antagonistas & inibidores , Humanos , Terapia de Imunossupressão , Imunoterapia/tendências , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Neoplasias/patologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Evasão Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA