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








Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 29(11): 2158-2169, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36951682

RESUMO

PURPOSE: G-CSF enhances colon cancer development. This study defines the prevalence and effects of increased G-CSF signaling in human colon cancers and investigates G-CSF inhibition as an immunotherapeutic strategy against metastatic colon cancer. EXPERIMENTAL DESIGN: Patient samples were used to evaluate G-CSF and G-CSF receptor (G-CSFR) levels by IHC with sera used to measure G-CSF levels. Peripheral blood mononuclear cells were used to assess the rate of G-CSFR+ T cells and IFNγ responses to chronic ex vivo G-CSF. An immunocompetent mouse model of peritoneal metastasis (MC38 cells in C57Bl/6J) was used to determine the effects of G-CSF inhibition (αG-CSF) on survival and the tumor microenvironment (TME) with flow and mass cytometry. RESULTS: In human colon cancer samples, the levels of G-CSF and G-CSFR are higher compared to normal colon tissues from the same patient. High patient serum G-CSF is associated with increases in markers of poor prognosis, (e.g., VEGF, IL6). Circulating T cells from patients express G-CSFR at double the rate of T cells from controls. Prolonged G-CSF exposure decreases T cell IFNγ production. Treatment with αG-CSF shifts both the adaptive and innate compartments of the TME and increases survival (HR, 0.46; P = 0.0237) and tumor T-cell infiltration, activity, and IFNγ response with greater effects in female mice. There is a negative correlation between serum G-CSF levels and tumor-infiltrating T cells in patient samples from women. CONCLUSIONS: These findings support G-CSF as an immunotherapeutic target against colon cancer with greater potential benefit in women.


Assuntos
Neoplasias do Colo , Fator Estimulador de Colônias de Granulócitos , Humanos , Feminino , Camundongos , Animais , Leucócitos Mononucleares , Linfócitos T , Receptores de Fator Estimulador de Colônias de Granulócitos/fisiologia , Neoplasias do Colo/tratamento farmacológico , Imunoterapia , Microambiente Tumoral
2.
Adv Sci (Weinh) ; 9(21): e2105868, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35599386

RESUMO

Immunogenic cell death (ICD) arises when cells are under stress, and their membranes are damaged. They release damage-associated molecular patterns (DAMPs) that stimulate and drive the type and magnitude of the immune response. In the presence of an antigen, DAMPs ride the longevity and efficacy of antigen-specific immunity. Yet, no tool can induce the controlled ICD with predictable results. A peptide-based tool, [II], is designed that aggregates in the cell and causes cell membrane damage, generates ICD and DAMPs release on various cell types, and hence can act as an adjuvant. An influenza vaccine is prepared by combining [II] with influenza hemagglutinin (HA) subunit antigens. The results show that [II] induced significantly higher HA-specific immunoglobulin G1 (IgG1) and IgG2a antibodies than HA-only immunized mice, while the peptide itself did not elicit antibodies. This paper demonstrates the first peptide-aggregation induced immunogenic rupture (PAIIR) approach as a vaccine adjuvant. PAIIR is a promising adjuvant with a high potential to promote universal protection upon influenza HA vaccination.


Assuntos
Vacinas contra Influenza , Influenza Humana , Adjuvantes Imunológicos , Animais , Anticorpos Antivirais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos
3.
J Am Soc Nephrol ; 33(4): 747-768, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35110364

RESUMO

BACKGROUND: Inducible disruption of cilia-related genes in adult mice results in slowly progressive cystic disease, which can be greatly accelerated by renal injury. METHODS: To identify in an unbiased manner modifier cells that may be influencing the differential rate of cyst growth in injured versus non-injured cilia mutant kidneys at a time of similar cyst severity, we generated a single-cell atlas of cystic kidney disease. We conducted RNA-seq on 79,355 cells from control mice and adult-induced conditional Ift88 mice (hereafter referred to as cilia mutant mice) that were harvested approximately 7 months post-induction or 8 weeks post 30-minute unilateral ischemia reperfusion injury. RESULTS: Analyses of single-cell RNA-seq data of CD45+ immune cells revealed that adaptive immune cells differed more in cluster composition, cell proportion, and gene expression than cells of myeloid origin when comparing cystic models with one another and with non-cystic controls. Surprisingly, genetic deletion of adaptive immune cells significantly reduced injury-accelerated cystic disease but had no effect on cyst growth in non-injured cilia mutant mice, independent of the rate of cyst growth or underlying genetic mutation. Using NicheNet, we identified a list of candidate cell types and ligands that were enriched in injured cilia mutant mice compared with aged cilia mutant mice and non-cystic controls that may be responsible for the observed dependence on adaptive immune cells during injury-accelerated cystic disease. CONCLUSIONS: Collectively, these data highlight the diversity of immune cell involvement in cystic kidney disease.


Assuntos
Cistos , Doenças Renais Policísticas , Animais , Cílios/metabolismo , Cistos/genética , Rim/metabolismo , Camundongos , Mutação , Doenças Renais Policísticas/metabolismo
4.
Theranostics ; 12(2): 639-656, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34976205

RESUMO

Rationale: B cells have emerged as key regulators in protective cancer immunity. However, the activation pathways induced in B cells during effective immunotherapy are not well understood. Methods: We used a novel localized ablative immunotherapy (LAIT), combining photothermal therapy (PTT) with intra-tumor delivery of the immunostimulant N-dihydrogalactochitosan (GC), to treat mice bearing mouse mammary tumor virus-polyoma middle tumor-antigen (MMTV-PyMT). We used single-cell RNA sequencing to compare the transcriptional changes induced by PTT, GC and PTT+GC in B cells within the tumor microenvironment (TME). Results: LAIT significantly increased survival in the tumor-bearing mice, compared to the treatment by PTT and GC alone. We found that PTT, GC and PTT+GC increased the proportion of tumor-infiltrating B cells and induced gene expression signatures associated with B cell activation. Both GC and PTT+GC elevated gene expression associated with antigen presentation, whereas GC elevated transcripts that regulate B cell activation and GTPase function and PTT+GC induced interferon response genes. Trajectory analysis, where B cells were organized according to pseudotime progression, revealed that both GC and PTT+GC induced the differentiation of B cells from a resting state towards an effector phenotype. The analyses confirmed upregulated interferon signatures in the differentiated tumor-infiltrating B cells following treatment by PTT+GC but not by GC. We also observed that breast cancer patients had significantly longer survival time if they had elevated expression of genes in B cells that were induced by PTT+GC therapy in the mouse tumors. Conclusion: Our findings show that the combination of local ablation and local application of immunostimulant initiates the activation of interferon signatures and antigen-presentation in B cells which is associated with positive clinical outcomes for breast cancer. These findings broaden our understanding of LAIT's regulatory roles in remodeling TME and shed light on the potentials of B cell activation in clinical applications.


Assuntos
Apresentação de Antígeno , Linfócitos B/imunologia , Imunoterapia , Interferons/metabolismo , Neoplasias Mamárias Experimentais/imunologia , Animais , Linfócitos B/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/mortalidade , Neoplasias Mamárias Experimentais/terapia , Camundongos , Transcriptoma
5.
J Infect Dis ; 225(8): 1494-1495, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-34929040
6.
J Infect Dis ; 225(5): 800-809, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-34865064

RESUMO

BACKGROUND: Coronavirus disease 2019 (COVID-19) disproportionally affects pregnant women and their newborn; however, little is known about variables that modulate maternal-fetal immune response to infection. METHODS: We prospectively studied socioeconomic, biologic, and clinical factors affecting humoral immunity in 87 unvaccinated pregnant women hospitalized in Buenos Aires for symptoms consistent with COVID-19. RESULTS: The number of days between symptom onset and childbirth predicted maternal and newborn virus spike protein receptor binding domain (RBD)-specific immunoglobulin G (IgG). These findings suggest newborns may benefit less when mothers deliver soon after COVID-19 infection. Similarly, a longer time between symptom onset and birth predicted higher in utero transfer of maternal IgG and its concentration in cord blood. Older gestational age at birth was associated with lower maternal to cord blood IgG ratio. Of women with confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, 87% developed RBD-specific IgA responses in breast milk within 96 hours of childbirth. IgA was not significantly associated with time from infection but correlated with maternal serum IgG and placental transfer. CONCLUSIONS: These results demonstrate the combined role of biologic, clinical, and socioeconomic variables associated with maternal RBD-specific antibodies and supports early vaccination strategies for COVID-19 in socioeconomically vulnerable pregnant women. CLINICAL TRIALS REGISTRATION: NCT04362956.


Assuntos
Anticorpos Antivirais/sangue , COVID-19/imunologia , COVID-19/transmissão , Imunoglobulina G/sangue , SARS-CoV-2/imunologia , Adulto , Produtos Biológicos , COVID-19/sangue , Teste Sorológico para COVID-19 , Feminino , Humanos , Recém-Nascido , Placenta/metabolismo , Gravidez , Estudos Prospectivos , Glicoproteína da Espícula de Coronavírus/imunologia , Populações Vulneráveis
7.
Infect Immun ; 89(11): e0043821, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34424751

RESUMO

All clinical Clostridioides difficile strains identified to date express a surface capsule-like polysaccharide structure known as polysaccharide II (PSII). The PSII antigen is immunogenic and, when conjugated to a protein carrier, induces a protective antibody response in animal models. Given that CD1d-restricted natural killer T (NKT) cells promote antibody responses, including those against carbohydrates, we tested the hypothesis that immunization with PSII and a CD1d-binding glycolipid adjuvant could lead to enhanced protection against a live C. difficile challenge. We purified PSII from a clinical isolate of C. difficile and immunized B6 mice with PSII alone or PSII plus the CD1d-binding glycolipid α-galactosylceramide (α-GC). PSII-specific IgM and IgG titers were evident in sera from immunized mice. The inclusion of α-GC had a modest influence on isotype switch but increased the IgG1/IgG2c ratio. Enhanced protection against C. difficile disease was achieved by inclusion of the α-GC ligand and was associated with reduced bacterial numbers in fecal pellets. In contrast, NKT-deficient Traj18-/- mice were not protected by the PSII/α-GC immunization modality. Absence of NKT cells similarly had a modest effect on isotype switch, but ratios of IgG1/IgG2c decreased. These results indicate that α-GC-driven NKT cells move the humoral immune response against C. difficile PSII antigen toward Th2-driven IgG1 and may contribute to augmented protection. This study suggests that NKT activation represents a pathway for additional B-cell help that could be used to supplement existing efforts to develop vaccines against polysaccharides derived from C. difficile and other pathogens.


Assuntos
Antígenos de Bactérias/imunologia , Clostridioides difficile/imunologia , Galactosilceramidas/imunologia , Imunoglobulina G/sangue , Células T Matadoras Naturais/imunologia , Polissacarídeos Bacterianos/imunologia , Animais , Anticorpos Antibacterianos/sangue , Feminino , Imunização , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL
8.
Infect Immun ; 89(10): e0027421, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34097471

RESUMO

The pathology associated with Clostridioides difficile disease is caused in large part by TcdB, an intracellular bacterial toxin that inactivates small GTPases. Despite C. difficile causing enteric disease, antitoxin IgG is a clear correlate of protection against infection-associated pathology. Immunization with TcdB-based immunogens or passive transfer of monoclonal antibodies specific for the TcdB carboxy-terminal domain (CTD) confers protection following C. difficile infection. Whether the mechanism by which circulating IgG is delivered to the gut depends on specific receptor-mediated transport or is solely reflective of infection-induced damage to the gut remains unclear. Here, we tested the hypothesis that neonatal Fc receptor (FcRn) is required for the delivery of systemic TcdB-specific IgG to the gut and protection against C. difficile-associated pathology. FcRn-expressing mice and FcRn-deficient littermates were immunized subcutaneously with Alhydrogel adjuvant-adsorbed CTD before challenge with live C. difficile spores. FcRn was required for the delivery of systemic TcdB-specific IgG to the gut and for vaccine-induced protection against C. difficile-associated disease. The lack of FcRn expression had minimal effects on the composition of the gut microbiome and did not affect susceptibility to C. difficile infection in nonimmunized mice. In further experiments, intraperitoneal injection of immune sera in FcRn-deficient mice led to the transport of protective IgG to the gut independently of infection, confirming a reported method of bypassing the FcRn. Our results reveal an FcRn-dependent mechanism by which systemic immunization-induced IgG protects the gut during enteric C. difficile infection. These findings may be beneficial for the targeting of C. difficile-specific IgG to the gut.


Assuntos
Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Sistema Digestório/imunologia , Sistema Digestório/microbiologia , Suscetibilidade a Doenças/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoglobulina G/imunologia , Receptores Fc/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Antitoxinas/imunologia , Toxinas Bacterianas/imunologia , Infecções por Clostridium/microbiologia , Suscetibilidade a Doenças/microbiologia , Enterotoxinas/imunologia , Feminino , Imunidade/imunologia , Imunização/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vacinação/métodos
9.
Front Immunol ; 12: 818734, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35095921

RESUMO

Adjuvant combinations may enhance or broaden the expression of immune responses to vaccine antigens. Information on whether established Alum type adjuvants can be combined with experimental CD1d ligand adjuvants is currently lacking. In this study, we used a murine Clostridioides difficile immunization and challenge model to evaluate Alum (Alhydrogel™), α-galactosylceramide (α-GC), and one of its analogs 7DW8-5 singly and in combination as vaccine adjuvants. We observed that the Alum/α-GC combination caused modest enhancement of vaccine antigen-specific IgG1 and IgG2b responses, and a broadening to include IgG2c that did not significantly impact overall protection. Similar observations were made using the Alum/7DW8-5 combination. Examination of the impact of adjuvants on NKT cells revealed expansion of invariant NKT (iNKT) cells with modest expansion of their iNKTfh subset and little effect on diverse NKT (dNKT) cells. Side effects of the adjuvants was determined and revealed transient hepatotoxicity when Alum/α-GC was used in combination but not singly. In summary these results showed that the Alum/α-GC or the Alum/7DW8-5 combination could exert distinct effects on the NKT cell compartment and on isotype switch to produce Th1-driven IgG subclasses in addition to Alum/Th2-driven subclasses. While Alum alone was efficacious in stimulating IgG-mediated protection, and α-GC offered no apparent additional benefit in the C. difficile challenge model, the work herein reveals immune response features that could be optimized and harnessed in other vaccine contexts.


Assuntos
Adjuvantes de Vacinas , Compostos de Alúmen , Vacinas Bacterianas/imunologia , Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Infecções por Clostridium/prevenção & controle , Células T Matadoras Naturais/imunologia , Adjuvantes de Vacinas/administração & dosagem , Adjuvantes de Vacinas/química , Animais , Vacinas Bacterianas/administração & dosagem , Biomarcadores , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Feminino , Imunização , Imunofenotipagem , Ligantes , Ativação Linfocitária/imunologia , Camundongos , Células T Matadoras Naturais/metabolismo
10.
Front Immunol ; 11: 565648, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042146

RESUMO

Bacterial enteric pathogens individually and collectively represent a serious global health burden. Humoral immune responses following natural or experimentally-induced infections are broadly appreciated to contribute to pathogen clearance and prevention of disease recurrence. Herein, we have compared observations on humoral immune mechanisms following infection with Citrobacter rodentium, the model for enteropathogenic Escherichia coli, Vibrio cholerae, Shigella species, Salmonella enterica species, and Clostridioides difficile. A comparison of what is known about the humoral immune responses to these pathogens reveals considerable variance in specific features of humoral immunity including establishment of high affinity, IgG class-switched memory B cell and long-lived plasma cell compartments. This article suggests that such variance could be contributory to persistent and recurrent disease.


Assuntos
Infecções Bacterianas/imunologia , Enteropatias/imunologia , Animais , Bactérias , Humanos , Imunidade Humoral
11.
JCI Insight ; 5(16)2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32663199

RESUMO

Clostridioides difficile is a leading cause of nosocomial infection responsible for significant morbidity and mortality with limited options for therapy. Secreted C. difficile toxin B (TcdB) is a major contributor to disease pathology, and select TcdB-specific Abs may protect against disease recurrence. However, the high frequency of recurrence suggests that the memory B cell response, essential for new Ab production following C. difficile reexposure, is insufficient. We therefore isolated TcdB-specific memory B cells from individuals with a history of C. difficile infection and performed single-cell deep sequencing of their Ab genes. Herein, we report that TcdB-specific memory B cell-encoded antibodies showed somatic hypermutation but displayed limited isotype class switch. Memory B cell-encoded mAb generated from the gene sequences revealed low to moderate affinity for TcdB and a limited ability to neutralize TcdB. These findings indicate that memory B cells are an important factor in C. difficile disease recurrence.


Assuntos
Anticorpos Neutralizantes/imunologia , Linfócitos B/imunologia , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Infecções por Clostridium/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Linfócitos B/microbiologia , Células CHO , Estudos de Casos e Controles , Clostridioides difficile/imunologia , Cricetulus , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Memória Imunológica , Pessoa de Meia-Idade , Hipermutação Somática de Imunoglobulina
12.
Infect Immun ; 88(3)2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-31871095

RESUMO

The intracellularly active bacterial toxin TcdB is a major Clostridioides difficile virulence factor that contributes to inflammation and tissue damage during disease. Immunization with an inactive TcdB fragment prevents C. difficile infection (CDI)-associated pathology. The protective immune response against inactive TcdB involves development of antigen-specific memory B cells and long-lived plasma cells that encode TcdB-neutralizing antibodies. Unlike the response to inactive TcdB, very little is known about the host humoral immune response to C. difficile and TcdB during primary and recurrent infection. Here, we used a murine model of C. difficile disease recurrence to demonstrate that an initial infection induced a serum IgM and mucosal IgA response against the toxin, but a low serum IgG response, which is associated with a lack of protection against disease during reinfection. Infection induced a partial expansion of the T follicular helper cell compartment, essential for B cell memory responses, and, consistent with that, failed to significantly expand the memory B cell compartment. Further, infection failed to stimulate the memory B cell compartment in preimmunized mice, although they were protected against associated disease. These results delineate the key humoral immune events that follow primary and recurrent C. difficile infection and provide a compelling inverse correlation between B cell memory and disease recurrence.


Assuntos
Linfócitos B/imunologia , Clostridioides difficile/imunologia , Infecções por Clostridium/imunologia , Imunização , Imunoglobulina G/sangue , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Toxinas Bacterianas/imunologia , Infecções por Clostridium/microbiologia , Imunoglobulina A/metabolismo , Camundongos , Mucosa/metabolismo
13.
Immunohorizons ; 3(3): 88-93, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-31342012

RESUMO

The CD1d-binding glycolipid α-galactosylceramide (α-GC) is a potent adjuvant that activates NKT cells and in turn enhances T-dependent humoral immunity. Very little is known about how NKT cells and the NKT follicular helper (NKTfh) subset influence the immune response to T-independent polysaccharides. In this study, we used a Cre-Lox approach to generate mice devoid of the Bcl6 master transcription factor in CD4 lineage cells and thus devoid of NKTfh cells but not total NKT cells. It was observed that α-GC-driven IgG1 class switch against a polysaccharide Ag was dependent on the NKTfh subset. However, α-GC was unable to stimulate a polysaccharide-specific Ab recall response. It was observed that NKT-derived IL-21 was able to exert limited influence on the IgG1 response and was therefore likely to work in concert with other factors. This work shows that α-GC-driven NKTfh cells can direct polysaccharide-specific B cell responses by promoting IgG1 class switch but do not provide signals needed for generation of polysaccharide-specific B cell memory.


Assuntos
Switching de Imunoglobulina/imunologia , Imunoglobulina G/imunologia , Memória Imunológica , Células T Matadoras Naturais/imunologia , Polissacarídeos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Comunicação Celular , Feminino , Galactosilceramidas/imunologia , Imunização , Switching de Imunoglobulina/genética , Imunoglobulina G/genética , Ativação Linfocitária , Masculino , Camundongos , Camundongos Transgênicos , Células T Matadoras Naturais/metabolismo , Linfócitos T Auxiliares-Indutores/metabolismo
14.
Infect Immun ; 87(8)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31138612

RESUMO

Clostridioides difficile toxin B (TcdB) is an intracellular toxin responsible for many of the pathologies of C. difficile infection. The two variant forms of TcdB (TcdB1 and TcdB2) share 92% sequence identity but have reported differences in rates of cell entry, autoprocessing, and overall toxicity. This 2,366-amino-acid, multidomain bacterial toxin glucosylates and inactivates small GTPases in the cytosol of target cells, ultimately leading to cell death. Successful cell entry and intoxication by TcdB are known to involve various conformational changes in the protein, including a proteolytic autoprocessing event. Previous studies found that amino acids 1753 to 1852 influence the conformational states of the proximal carboxy-terminal domain of TcdB and could contribute to differences between TcdB1 and TcdB2. In the current study, a combination of approaches was used to identify sequences within the region from amino acids 1753 to 1852 that influence the conformational integrity and cytotoxicity of TcdB2. Four deletion mutants with reduced cytotoxicity were identified, while one mutant, TcdB2Δ1769-1787, exhibited no detectable cytotoxicity. TcdB2Δ1769-1787 underwent spontaneous autoprocessing and was unable to interact with CHO-K1 or HeLa cells, suggesting a potential change in the conformation of the mutant protein. Despite the putative alteration in structural stability, vaccination with TcdB2Δ1769-1787 induced a TcdB2-neutralizing antibody response and protected against C. difficile disease in a mouse model. These findings indicate that the 19-amino-acid region spanning residues 1769 to 1787 in TcdB2 is crucial to cytotoxicity and the structural regulation of autoprocessing and that TcdB2Δ1769-1787 is a promising candidate for vaccination.


Assuntos
Proteínas de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Clostridioides difficile/imunologia , Proteínas Repressoras/imunologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Células CHO , Cricetulus , Glicosilação , Células HeLa , Humanos , Camundongos , Conformação Proteica , Domínios Proteicos , Proteínas Repressoras/química , Proteínas Repressoras/fisiologia , Deleção de Sequência , Vacinação
15.
Clin Cancer Res ; 24(21): 5335-5346, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30068705

RESUMO

Purpose: To develop a synergistic combination therapy for advanced pancreatic cancer, using local phototherapy and immunotherapy, and to determine the efficacy and mechanism of the novel combination therapy using a highly metastatic pancreatic tumor model in mice.Experimental Design: Mice bearing Panc02-H7 pancreatic tumors (both subcutaneous and orthotopic) were treated with noninvasive or interventional photothermal therapy, followed by local application of an immunoadjuvant. Tumor growth and animal survival were assessed. Immune cell populations within spleen and tumors were evaluated by FACS and IHC, and cytokine levels were determined by ELISA.Results: Up to 75% of mice bearing subcutaneous tumors treated with combination therapy had complete tumor regression. Local photothermal therapy exposed/released damage-associated molecular patterns, which initiated an immunogenic tumor cell death, resulting in infiltration of antigen-presenting cells and Th1 immunity. Concomitant application of immunoadjuvant amplified Th1 immunity, especially the tumor-specific cytotoxic T lymphocyte response, with increased quantity and quality of T cells. Combination therapy also induced tumor-specific immune memory, as demonstrated by resistance to tumor rechallenge and production of memory T cells. For the treatment of orthotopic tumor, the combination therapy significantly reduced the primary tumors and metastases, and prolonged the animal survival time.Conclusions: This study indicated that combination of local phototherapy and immunotherapy induced a systemic immunity against established tumors and metastases in an aggressive, preclinical pancreatic tumor model, leading to a potential clinical method for patients with advanced pancreatic cancer. Clin Cancer Res; 24(21); 5335-46. ©2018 AACR.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas Anticâncer/imunologia , Imunogenicidade da Vacina , Neoplasias Pancreáticas/imunologia , Fotoquimioterapia , Animais , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular , Terapia Combinada , Modelos Animais de Doenças , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Camundongos , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Fotoquimioterapia/métodos , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Front Immunol ; 9: 305, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29520280

RESUMO

Vaccination with CD1d-binding glycolipid adjuvants and co-administered protein, lipid, and carbohydrate antigens leads to invariant natural killer T (NKT) cell-dependent enhancement of protective B cell responses. NKT cell activation boosts the establishment of protein antigen-specific B cell memory and long-lived plasma cell (LLPC) compartments. NKT cells may exert a similar effect on some carbohydrate-specific B cells, but not lipid-specific B cells. The mechanisms of action of NKT cells on B cell responsiveness and subsequent differentiation into memory B cells and LLPC is dependent on CD1d expression by dendritic cells and B cells that can co-present glycolipids on CD1d and antigen-derived peptide on MHCII. CD1d/glycolipid-activated NKT cells are able to provide help to B cells in a manner dependent on cognate and non-cognate interactions. More recently, a glycolipid-expanded subset of IL-21-secreting NKT cells known as NKT follicular helper cells has been suggested to be a driver of NKT-enhanced humoral immunity. This review summarizes established and recent findings on how NKT cells impact humoral immunity and suggests possible areas of investigation that may allow the incorporation of NKT-activating agents into vaccine adjuvant platforms.


Assuntos
Antígenos/imunologia , Imunidade Humoral , Memória Imunológica , Ativação Linfocitária , Células T Matadoras Naturais/imunologia , Plasmócitos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Humanos
17.
Front Immunol ; 9: 3064, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30697210

RESUMO

Memory B cells that are generated during an infection or following vaccination act as sentinels to guard against future infections. Upon repeat antigen exposure memory B cells differentiate into new antibody-secreting plasma cells to provide rapid and sustained protection. Some pathogens evade or suppress the humoral immune system, or induce memory B cells with a diminished ability to differentiate into new plasma cells. This leaves the host vulnerable to chronic or recurrent infections. Single cell approaches coupled with next generation antibody gene sequencing facilitate a detailed analysis of the pathogen-specific memory B cell repertoire. Monoclonal antibodies that are generated from antibody gene sequences allow a functional analysis of the repertoire. This review discusses what has been learned thus far from analysis of diverse pathogen-specific memory B cell compartments and describes major differences in their repertoires. Such information may illuminate ways to advance the goal of improving vaccine and therapeutic antibody design.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Memória Imunológica , Plasmócitos/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Antígenos/imunologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Humoral/imunologia , Plasmócitos/metabolismo
19.
Blood ; 129(26): 3398-3399, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28663222
20.
Immunology ; 152(1): 36-51, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28419443

RESUMO

The role of the unique T-cell population, natural killer T (NKT) cells, which have similar functions to NK cells in pancreatic cancer (PC), is not yet evaluated. To address the regulatory roles of NKT cells on tumour progression through tumour-associated macrophages (TAM) and their production of microsomal prostaglandin E synthase-1 (mPGES-1) and 5-lipoxygenase (5-LOX) in (Kras)-driven pancreatic tumour (KPT) progression, we crossed CD1d-/- mice deficient in both invariant and variant NKT cells with the KrasG12D mice. Loss of NKT cells significantly increased pancreatic intraepithelial neoplasia (PanIN) lesions and also increased 5-LOX and mPGES-1 expression in M2-type macrophages and cancer stem-like cells in pancreatic tumours. Pharmacological inhibition of mPGES-1 and 5-LOX in M2 macrophages with specific inhibitor YS-121 in KPT-CD1d-/- mice decreased PanIN lesions and suppressed tumour growth in association with elevated levels of active CD8a cells. Hence, NKT cells regulate PC by modulating TAMs (M2) through mPGES-1 and 5-LOX; and the absence of NKT cells leads to aggressive development of PC.


Assuntos
Carcinoma in Situ/imunologia , Macrófagos/imunologia , Células T Matadoras Naturais/imunologia , Neoplasias Pancreáticas/imunologia , Animais , Antígenos CD1d/genética , Araquidonato 5-Lipoxigenase/imunologia , Araquidonato 5-Lipoxigenase/metabolismo , Antígenos CD8/imunologia , Antígenos CD8/metabolismo , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Carcinoma in Situ/prevenção & controle , Proliferação de Células , Progressão da Doença , Genes ras , Predisposição Genética para Doença , Humanos , Inibidores de Lipoxigenase/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/prevenção & controle , Fenótipo , Prostaglandina-E Sintases/antagonistas & inibidores , Prostaglandina-E Sintases/imunologia , Prostaglandina-E Sintases/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA