Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(5): 1055-1065.e5, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33945786

RESUMEN

Efforts are being made worldwide to understand the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for the coronavirus disease 2019 (COVID-19) pandemic, including the impact of T cell immunity and cross-recognition with seasonal coronaviruses. Screening of SARS-CoV-2 peptide pools revealed that the nucleocapsid (N) protein induced an immunodominant response in HLA-B7+ COVID-19-recovered individuals that was also detectable in unexposed donors. A single N-encoded epitope that was highly conserved across circulating coronaviruses drove this immunodominant response. In vitro peptide stimulation and crystal structure analyses revealed T cell-mediated cross-reactivity toward circulating OC43 and HKU-1 betacoronaviruses but not 229E or NL63 alphacoronaviruses because of different peptide conformations. T cell receptor (TCR) sequencing indicated that cross-reactivity was driven by private TCR repertoires with a bias for TRBV27 and a long CDR3ß loop. Our findings demonstrate the basis of selective T cell cross-reactivity for an immunodominant SARS-CoV-2 epitope and its homologs from seasonal coronaviruses, suggesting long-lasting protective immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Epítopos Inmunodominantes/inmunología , SARS-CoV-2/inmunología , Secuencia de Aminoácidos , Coronavirus/clasificación , Coronavirus/inmunología , Proteínas de la Nucleocápside de Coronavirus/química , Reacciones Cruzadas , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Antígeno HLA-B7/química , Antígeno HLA-B7/genética , Antígeno HLA-B7/inmunología , Humanos , Epítopos Inmunodominantes/química , Memoria Inmunológica , Modelos Moleculares , Péptidos/química , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
2.
Br J Haematol ; 204(2): 415-433, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38155519

RESUMEN

Epstein-Barr virus (EBV)-associated lymphomas cover a range of histological B- and T-cell non-Hodgkin and Hodgkin lymphoma subtypes. The role of EBV on B-cell malignant pathogenesis and its impact on the tumour microenvironment are intriguing but incompletely understood. Both the International Consensus Classification (ICC) and 5th Edition of the World Health Organization (WHO-HAEM5) proposals give prominence to the distinct clinical, prognostic, genetic and tumour microenvironmental features of EBV in lymphoproliferative disorders. There have been major advances in our biological understanding, in how to harness features of EBV and its host immune response for targeted therapy, and in using EBV as a method to monitor disease response. In this article, we showcase the latest developments and how they may be integrated to stimulate new and innovative approaches for further lines of investigation and therapy.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Enfermedad de Hodgkin , Linfoma no Hodgkin , Linfoma , Trastornos Linfoproliferativos , Humanos , Herpesvirus Humano 4/genética , Enfermedad de Hodgkin/patología , Microambiente Tumoral
3.
PLoS Pathog ; 18(6): e1010403, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35737741

RESUMEN

There is now convincing evidence that the successful development of an effective CMV vaccine will require improved formulation and adjuvant selection that is capable of inducing both humoral and cellular immune responses. Here, we have designed a novel bivalent subunit vaccine formulation based on CMV-encoded oligomeric glycoprotein B (gB) and polyepitope protein in combination with human compatible TLR9 agonist CpG1018. The polyepitope protein includes multiple minimal HLA class I-restricted CD8+ T cell epitopes from different antigens of CMV. This subunit vaccine generated durable anti-viral antibodies, CMV-specific CD4+ and CD8+ T cell responses in multiple HLA expressing mice. Antibody responses included broad TH1 isotypes (IgG2a, IgG2b and IgG3) and potently neutralized CMV infection in fibroblasts and epithelial cells. Furthermore, polyfunctional antigen-specific T cell immunity and antiviral antibody responses showed long-term memory maintenance. These observations argue that this novel vaccine strategy, if applied to humans, could facilitate the generation of robust humoral and cellular immune responses which may be more effective in preventing CMV-associated complications in various clinical settings.


Asunto(s)
Infecciones por Citomegalovirus , Inmunidad Humoral , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales , Linfocitos T CD8-positivos , Epítopos de Linfocito T/metabolismo , Humanos , Inmunidad Celular , Ratones , Oligodesoxirribonucleótidos , Receptor Toll-Like 9/metabolismo , Vacunas Combinadas , Vacunas de Subunidad
4.
PLoS Pathog ; 18(2): e1010339, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35157735

RESUMEN

Adoptive T-cell immunotherapy has provided promising results in the treatment of viral complications in humans, particularly in the context of immunocompromised patients who have exhausted all other clinical options. The capacity to expand T cells from healthy immune individuals is providing a new approach to anti-viral immunotherapy, offering rapid off-the-shelf treatment with tailor-made human leukocyte antigen (HLA)-matched T cells. While most of this research has focused on the treatment of latent viral infections, emerging evidence that SARS-CoV-2-specific T cells play an important role in protection against COVID-19 suggests that the transfer of HLA-matched allogeneic off-the-shelf virus-specific T cells could provide a treatment option for patients with active COVID-19 or at risk of developing COVID-19. We initially screened 60 convalescent individuals and based on HLA typing and T-cell response profile, 12 individuals were selected for the development of a SARS-CoV-2-specific T-cell bank. We demonstrate that these T cells are specific for up to four SARS-CoV-2 antigens presented by a broad range of both HLA class I and class II alleles. These T cells show consistent functional and phenotypic properties, display cytotoxic potential against HLA-matched targets and can recognize HLA-matched cells infected with different SARS-CoV-2 variants. These observations demonstrate a robust approach for the production of SARS-CoV-2-specific T cells and provide the impetus for the development of a T-cell repository for clinical assessment.


Asunto(s)
Antígenos HLA/inmunología , Inmunoterapia Adoptiva , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Adulto , Epítopos de Linfocito T , Femenino , Células HEK293 , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Angiogenesis ; 25(2): 181-203, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34617194

RESUMEN

Several B-cell subsets with distinct functions and polarized cytokine profiles that extend beyond antibody production have been reported in different cancers. Here we have demonstrated that proliferating B cells were predominantly found in the peritumoral region of esophageal squamous cell carcinoma (ESCC). These B cells were enriched in tumor nests with high expression of high-mobility group box 1 (HMGB1). High densities of peritumoral proliferating B cells and concomitantly high intratumoral HMGB1 expression showed improved prognostic significance, surpassing prognostic stratification of ESCC patients based on HMGB1 positivity alone. This striking association led us to set up models to test whether cancer-derived HMGB1 could shape tumor microenvironment via modulation on B cells. Overexpression of HMGB1 in ESCC cell lines (KYSE510 and EC18) enhanced proliferation and migration of B cells. Transcriptomic analysis showed that migratory B cells exhibited high enrichment of proangiogenic genes. VEGF expression in proliferating B cells was induced upon co-culture of HMGB1-overexpressing tumor cells and B cells. Secretome array profiling of conditioned media (CM) from the co-culture revealed rich expression of proangiogenic proteins. Consequently, incubation of human umbilical vein endothelial cells with CM promoted angiogenesis in tube formation and migration assays. HMGB1 inhibitor, glycyrrhizin, abolishes all the observed proangiogenic phenotypes. Finally, co-injection of B cells and CM with HMGB1-overexpressing tumor cells, but not with glycyrrhizin, significantly enhanced tumor growth associated with increased microvascular density in ESCC xenograft mice model. Our results indicate that cancer-derived HMGB1 elevates angiogenesis in ESCC by shifting the balance toward proangiogenic signals in proliferating B cells.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Proteína HMGB1 , Animales , Línea Celular Tumoral , Proliferación Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Ácido Glicirrínico , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Neovascularización Patológica/patología , Microambiente Tumoral
6.
J Virol ; 95(6)2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33443088

RESUMEN

Developing optimal T-cell response assays to severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is critical for measuring the duration of immunity to this disease and assessing the efficacy of vaccine candidates. These assays need to target conserved regions of SARS-CoV-2 global variants and avoid cross-reactivity to seasonal human coronaviruses. To contribute to this effort, we employed an in silico immunoinformatics analysis pipeline to identify immunogenic peptides resulting from conserved and highly networked regions with topological importance from the SARS-CoV-2 nucleocapsid and spike proteins. A total of 57 highly networked T-cell epitopes that are conserved across geographic viral variants were identified from these viral proteins, with a binding potential to diverse HLA alleles and 80 to 100% global population coverage. Importantly, 18 of these T-cell epitope derived peptides had limited homology to seasonal human coronaviruses making them promising candidates for SARS-CoV-2-specific T-cell immunity assays. Moreover, two of the NC-derived peptides elicited effector/polyfunctional responses of CD8+ T cells derived from SARS-CoV-2 convalescent patients.IMPORTANCE The development of specific and validated immunologic tools is critical for understanding the level and duration of the cellular response induced by SARS-CoV-2 infection and/or vaccines against this novel coronavirus disease. To contribute to this effort, we employed an immunoinformatics analysis pipeline to define 57 SARS-CoV-2 immunogenic peptides within topologically important regions of the nucleocapsid (NC) and spike (S) proteins that will be effective for detecting cellular immune responses in 80 to 100% of the global population. Our immunoinformatics analysis revealed that 18 of these peptides had limited homology to circulating seasonal human coronaviruses and therefore are promising candidates for distinguishing SARS-CoV-2-specific immune responses from pre-existing coronavirus immunity. Importantly, CD8+ T cells derived from SARS-CoV-2 survivors exhibited polyfunctional effector responses to two novel NC-derived peptides identified as HLA-binders. These studies provide a proof of concept that our immunoinformatics analysis pipeline identifies novel immunogens which can elicit polyfunctional SARS-CoV-2-specific T-cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Epítopos de Linfocito T/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Secuencia de Aminoácidos , Presentación de Antígeno , COVID-19/sangre , COVID-19/inmunología , Biología Computacional , Coronavirus/clasificación , Coronavirus/inmunología , Proteínas de la Nucleocápside de Coronavirus/química , Proteínas de la Nucleocápside de Coronavirus/genética , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Antígenos HLA/inmunología , Humanos , Inmunidad Celular , Mutación , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Unión Proteica , SARS-CoV-2/genética , Especificidad de la Especie , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética
7.
J Infect Dis ; 224(2): 312-317, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-33274385

RESUMEN

Cytomegalovirus (CMV) remains a significant burden in lung transplant recipients. Deficiencies in T-cell immunity posttransplant increase the risk of CMV-associated complications. However, it is not clear if underlying poor pretransplant immunity increases risk. To assess this, we recruited 39 prospective lung transplant patients and performed QuantiFERON-CMV on their peripheral blood. More than a third of prospective CMV-seropositive transplant recipients were CMV non-immune reactive (CMV-NIR) pretransplant. CMV-NIR status was associated with a significantly higher incidence of CMV reactivation posttransplant, demonstrating that dysfunctional CMV immunity in prospective lung transplant recipients is associated with an increased risk of viral reactivation posttransplant.


Asunto(s)
Infecciones por Citomegalovirus , Inmunidad Celular , Trasplante de Pulmón , Infecciones por Citomegalovirus/epidemiología , Infecciones por Citomegalovirus/inmunología , Humanos , Infección Latente/virología , Trasplante de Pulmón/efectos adversos , Estudios Prospectivos
9.
Int J Cancer ; 146(8): 2305-2314, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-31950498

RESUMEN

Now is an exciting era of development in immunotherapy checkpoint inhibitors and their effect on the treatment of NPC. While the general prognosis of R/M disease is poor, immunotherapy offers some promise in a malignancy associated with EBV and characterized by a peritumoural immune infiltrate. Our study aims to review past and on-going clinical trials of monoclonal antibody therapies against the checkpoint inhibitors (e.g. PD1 and CTLA-4), in R/M NPC. All randomized and nonrandomized controlled trials involving immune checkpoint inhibitor interventions for treatment of NPC were included in the study. We utilized a validated "risk of bias" tool to assess study quality. Four separate Phase I-II trials report the potential of PD1 inhibitor treatment for patients with NPC. Within the observed groups, camrelizumab combined with chemotherapy achieved an objective response in 91% of patients as first-line treatment for metastatic NPC (PFS 68% at 1-year) but this was associated with a high rate of grade >3 adverse events (87%; CTCAE version 4.03). The remaining three studies focused on recurrent NPC disease in patients who had received at least one line of prior chemotherapy. Within this group, camrelizumab monotherapy achieved an objective response in 34% of patients (PFS 27% at 1-year; range across all three studies 20.5-34%). No NPC trial has yet reported on specific outcomes for non-PD1 checkpoint inhibitors but 11 on-going studies include alternative targets (e.g. PD-L1/CTLA-4) as combination or monotherapy treatments. In considering checkpoint immunotherapies for NPC, initial results show promise for anti-PD1 interventions. Further phase I-III trials are in progress to clarify clinical outcomes, fully determine safety profiles, and optimize drug combinations and administration schedules.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno CTLA-4/antagonistas & inhibidores , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno B7-H1/inmunología , Antígeno CTLA-4/inmunología , Quimioradioterapia , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Humanos , Carcinoma Nasofaríngeo/inmunología , Neoplasias Nasofaríngeas/inmunología , Nivolumab/administración & dosificación , Nivolumab/uso terapéutico , Receptor de Muerte Celular Programada 1/inmunología
10.
Transpl Infect Dis ; 22(6): e13399, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32608543

RESUMEN

INTRODUCTION: BK virus (BKPyV) nephropathy occurs in 1%-10% of kidney transplant recipients, with suboptimal therapeutic options. CASE: A 54-year-old woman received a transplant in March 2017. BKPyV was detected at 1.5 × 102  copies/mL within a month, necessitating halving of mycophenolate and addition of leflunomide. Allograft histology in December showed polyomavirus nephropathy treated with intravenous immunoglobulin and cessation of mycophenolate. In February 2018, cidofovir and ciprofloxacin were commenced. In April, tacrolimus was reduced while introducing everolimus. A second graft biopsy in August showed increasing polyoma virus infection and a subsequent biopsy in September for worsening renal function showed 30% of tubular reactivity for simian virus 40 (SV40). Allogeneic BKPyV-reactive T cells were generated from the patient's daughter and infused over 10 sessions starting late September. The fourth allograft biopsy in November 2018 demonstrated involvement of BKPyV in 50% of tubules. Allograft function continued to decline, requiring hemodialysis from December 2018. Allograft nephrectomy after 6 months showed <1% SV40 in preserved tubules and 80% interstitial fibrosis. DISCUSSION: We conclude that the T-cell adoptive immunotherapy reduced BKPyV load significantly despite extensive infection, but attendant fibrosis and tubular atrophy led to graft failure. Early intervention with T-cell therapy may prove efficacious in BKPyV nephropathy.


Asunto(s)
Virus BK , Inmunoterapia Adoptiva , Trasplante de Riñón , Infecciones por Polyomavirus , Infecciones Tumorales por Virus , Australia , Femenino , Humanos , Leucocitos Mononucleares , Persona de Mediana Edad , Infecciones por Polyomavirus/terapia , Linfocitos T , Infecciones Tumorales por Virus/terapia
11.
Clin Infect Dis ; 68(4): 632-640, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982441

RESUMEN

Background: Opportunistic infections including cytomegalovirus (CMV) are a major cause of morbidity and mortality in solid organ transplant (SOT) recipients. The recurrent and protracted use of antiviral drugs with eventual emergence of drug resistance represents a significant constraint to therapy. Although adoptive T-cell therapy has been successfully used in hematopoietic stem cell transplant recipients, its extension to the SOT setting poses a considerable challenge because of the inhibitory effects of immunosuppressive drugs on the virus-specific T-cell response in vivo and the perceived risk of graft rejection. Methods: In this prospective study, 22 SOT recipients (13 renal and 8 lung and 1 heart transplants) with recurrent or ganciclovir-resistant CMV infection were recruited, and 13 of them were treated with in vitro-expanded autologous CMV-specific T cells. These patients were monitored for safety, clinical symptoms, and immune reconstitution. Results: Autologous CMV-specific T-cell manufacture was attempted for 21 patients, and was successful in 20. The use of this adoptive immunotherapy was associated with no therapy-related serious adverse events. Eleven (84%) of the 13 treated patients showed improvement in symptoms, including complete resolution or reduction in DNAemia and CMV-associated end-organ disease and/or the cessation or reduced use of antiviral drugs. Furthermore, four of these patients showed coincident increased frequency of CMV-specific T cells in peripheral blood after completion of T-cell therapy. Conclusions: The data presented here demonstrate for the first time the clinical safety of CMV-specific adoptive T-cell therapy and its potential therapeutic benefit for SOT recipients with recurrent and/or drug-resistant CMV infection or disease. Clinical Trials Registration: ACTRN12613000981729.


Asunto(s)
Traslado Adoptivo/métodos , Infecciones por Citomegalovirus/terapia , Citomegalovirus/inmunología , Linfocitos T/inmunología , Trasplante Autólogo/métodos , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Recurrencia , Resultado del Tratamiento , Adulto Joven
12.
Clin Microbiol Rev ; 30(2): 503-528, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28298471

RESUMEN

BK polyomavirus (BKV) causes frequent infections during childhood and establishes persistent infections within renal tubular cells and the uroepithelium, with minimal clinical implications. However, reactivation of BKV in immunocompromised individuals following renal or hematopoietic stem cell transplantation may cause serious complications, including BKV-associated nephropathy (BKVAN), ureteric stenosis, or hemorrhagic cystitis. Implementation of more potent immunosuppression and increased posttransplant surveillance has resulted in a higher incidence of BKVAN. Antiviral immunity plays a crucial role in controlling BKV replication, and our increasing knowledge about host-virus interactions has led to the development of improved diagnostic tools and clinical management strategies. Currently, there are no effective antiviral agents for BKV infection, and the mainstay of managing reactivation is reduction of immunosuppression. Development of immune-based therapies to combat BKV may provide new and exciting opportunities for the successful treatment of BKV-associated complications.


Asunto(s)
Virus BK/inmunología , Infecciones por Polyomavirus , Infecciones Tumorales por Virus , Antivirales , Virus BK/genética , Humanos , Huésped Inmunocomprometido , Infecciones por Polyomavirus/diagnóstico , Infecciones por Polyomavirus/inmunología , Infecciones por Polyomavirus/terapia , Infecciones por Polyomavirus/virología , Infecciones Tumorales por Virus/diagnóstico , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/terapia , Infecciones Tumorales por Virus/virología
13.
J Clin Microbiol ; 56(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29343541

RESUMEN

Immune reconstitution following organ transplantation is absolutely critical in preventing infectious complications. However, understanding the kinetics of immune reconstitution and its potential impact on the clinical management of transplant patients remains a significant challenge. Over the last decade, various platform technologies have emerged which have provided important insights into the immune reconstitution kinetics in transplant patients. However, many of these technologies are too complicated and cumbersome to implement in a clinical setting. In this issue of the Journal of Clinical Microbiology, Chiereghin et al. (J. Clin. Microbiol. 56:e01040-17, 2018, https://doi.org/10.1128/JCM.01040-17) report the results of their evaluation of the QuantiFERON-CMV (QFN-CMV) assay to assess human cytomegalovirus (CMV)-specific CD8+ T-cell immunity in heart transplant recipients as a prognostic tool. These studies showed that patients with absence of global immune reactivity in the QFN-CMV assay were at a higher risk of developing CMV after discontinuing antiviral prophylaxis. Furthermore, failure to reconstitute CMV-specific immunity after resolution of the first episode of viremia was associated with viral relapse. These observations, along with other recent clinical studies utilizing the QFN-CMV assay, demonstrate that systematic monitoring of antiviral immunity can be successfully used as a prognostic tool and also to guide changes to the clinical management of transplant patients.


Asunto(s)
Antivirales , Infecciones por Citomegalovirus , Trasplante de Corazón , Citomegalovirus , Humanos , Inmunidad Celular , Monitorización Inmunológica
14.
PLoS Pathog ; 12(8): e1005799, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27540722

RESUMEN

Disruption of T cell memory during severe immune suppression results in reactivation of chronic viral infections, such as Epstein Barr virus (EBV) and Cytomegalovirus (CMV). How different subsets of memory T cells contribute to the protective immunity against these viruses remains poorly defined. In this study we examined the compartmentalization of virus-specific, tissue resident memory CD8+ T cells in human lymphoid organs. This revealed two distinct populations of memory CD8+ T cells, that were CD69+CD103+ and CD69+CD103-, and were retained within the spleen and tonsils in the absence of recent T cell stimulation. These two types of memory cells were distinct not only in their phenotype and transcriptional profile, but also in their anatomical localization within tonsils and spleen. The EBV-specific, but not CMV-specific, CD8+ memory T cells preferentially accumulated in the tonsils and acquired a phenotype that ensured their retention at the epithelial sites where EBV replicates. In vitro studies revealed that the cytokine IL-15 can potentiate the retention of circulating effector memory CD8+ T cells by down-regulating the expression of sphingosine-1-phosphate receptor, required for T cell exit from tissues, and its transcriptional activator, Kruppel-like factor 2 (KLF2). Within the tonsils the expression of IL-15 was detected in regions where CD8+ T cells localized, further supporting a role for this cytokine in T cell retention. Together this study provides evidence for the compartmentalization of distinct types of resident memory T cells that could contribute to the long-term protection against persisting viral infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Herpesvirus Humano 4/inmunología , Memoria Inmunológica , Antígenos CD/inmunología , Linfocitos T CD8-positivos/patología , Infecciones por Citomegalovirus/patología , Infecciones por Virus de Epstein-Barr/patología , Femenino , Humanos , Interleucina-15/inmunología , Factores de Transcripción de Tipo Kruppel/inmunología , Masculino , Especificidad de Órganos/inmunología
16.
Immunol Cell Biol ; 95(4): 364-371, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27999434

RESUMEN

Persistent viral infections are associated with the majority of human cancers where infectious agents have been recognized as the primary etiological agent. These viruses contribute to the malignant transformation of human cells either through the expression of oncogenic proteins or chronic inflammation. In spite of the high prevalence of these viral infections in humans, only a small proportion of these individuals who may have an underlying immune defect develop malignant disease. Furthermore, many of these viruses have evolved unique mechanisms to avoid the host immune system to successfully establish latent infection with limited gene expression. Technological advances in delineating the role of cellular immune responses in the control of viral infections and ability to rapidly expand these effector cells in vitro have provided an important platform for the development of novel immunotherapeutic strategies to treat virus-associated cancers. While autologous T cell therapies have provided promising results, development of 'off-the-shelf' third-party allogeneic virus-specific T cell therapies have emerged as powerful tools to treat many of the virus-associated diseases. It is anticipated that adoptive T cell therapy in combination with newly emerging immune checkpoint inhibitors and therapeutic vaccines will provide opportunities to successfully treat advanced metastatic virus-associated cancers which are currently not amenable to standard therapeutic strategies.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias/prevención & control , Virus Oncogénicos/fisiología , Linfocitos T/inmunología , Infecciones Tumorales por Virus/terapia , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Antígenos Virales/inmunología , Transformación Celular Neoplásica , Terapia Combinada , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Humanos , Evasión Inmune , Neoplasias/etiología , Neoplasias/inmunología , Medicina de Precisión , Linfocitos T/trasplante , Infecciones Tumorales por Virus/inmunología , Latencia del Virus
17.
J Virol ; 90(16): 7497-507, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27279616

RESUMEN

UNLABELLED: Reconstitution of T cell immunity is absolutely critical for the effective control of virus-associated infectious complications in hematopoietic stem cell transplant (HSCT) recipients. Coinfection with genetic variants of human cytomegalovirus (CMV) in transplant recipients has been linked to clinical disease manifestation; however, how these genetic variants impact T cell immune reconstitution remains poorly understood. In this study, we have evaluated dynamic changes in the emergence of genetic variants of CMV in HSCT recipients and correlated these changes with reconstitution of antiviral T cell responses. In an analysis of single nucleotide polymorphisms within sequences encoding HLA class I-restricted CMV epitopes from the immediate early 1 gene of CMV, coinfection with genetically distinct variants of CMV was detected in 52% of patients. However, in spite of exposure to multiple viral variants, the T cell responses in these patients were preferentially directed to a limited repertoire of HLA class I-restricted CMV epitopes, either conserved, variant, or cross-reactive. More importantly, we also demonstrate that long-term control of CMV infection after HSCT is primarily mediated through the efficient induction of stable antiviral T cell immunity irrespective of the nature of the antigenic target. These observations provide important insights for the future design of antiviral T cell-based immunotherapeutic strategies for transplant recipients, emphasizing the critical impact of robust immune reconstitution on efficient control of viral infection. IMPORTANCE: Infection and disease caused by human cytomegalovirus (CMV) remain a significant burden in patients undergoing hematopoietic stem cell transplantation (HSCT). The establishment of efficient immunological control, primarily mediated by cytotoxic T cells, plays a critical role in preventing CMV-associated disease in transplant recipients. Recent studies have also begun to investigate the impact genetic variation in CMV has upon disease outcome in transplant recipients. In this study, we sought to investigate the role T cell immunity plays in recognizing and controlling genetic variants of CMV. We demonstrate that while a significant proportion of HSCT recipients may be exposed to multiple genetic variants of CMV, this does not necessarily lead to immune control mediated via recognition of this genetic variation. Rather, immune control is associated with the efficient establishment of a stable immune response predominantly directed against immunodominant conserved T cell epitopes.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Citomegalovirus/inmunología , Genotipo , Trasplante de Células Madre Hematopoyéticas , Linfocitos T/inmunología , Receptores de Trasplantes , Coinfección/inmunología , Coinfección/virología , Citomegalovirus/clasificación , Citomegalovirus/genética , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Genes Inmediatos-Precoces , Humanos , Polimorfismo de Nucleótido Simple
18.
Cancer Immunol Immunother ; 65(2): 213-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26759008

RESUMEN

Spread of head and neck cancer along the cranial nerves is often a lethal complication of this tumour. Current treatment options include surgical resection and/or radiotherapy, but recurrence is a frequent event suggesting that our understanding of this tumour and its microenvironment is incomplete. In this study, we have analysed the nature of the perineural tumour microenvironment by immunohistochemistry with particular focus on immune cells and molecules, which might impair anti-tumour immunity. Moderate to marked lymphocyte infiltrates were present in 58.8% of the patient cohort including T cells, B cells and FoxP3-expressing T cells. While human leukocyte antigen (HLA) class I and more variably HLA class II were expressed on the tumour cells, this did not associate with patient survival or recurrence. In contrast, galectin-1 staining within lymphocyte areas of the tumour was significantly associated with a poorer patient outcome. Given the known role of galectin-1 in immune suppression, the data suggest that galectin inhibitors might improve the prognosis of patients with perineural spread of cancer.


Asunto(s)
Galectina 1/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/mortalidad , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/mortalidad , Anciano , Nervios Craneales/patología , Femenino , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Estimación de Kaplan-Meier , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Subgrupos Linfocitarios/patología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Recurrencia Local de Neoplasia , Evaluación del Resultado de la Atención al Paciente , Pronóstico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Microambiente Tumoral
19.
Immunol Cell Biol ; 94(10): 964-974, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27297581

RESUMEN

Cross-presentation of exogenous protein antigens by B cells through the major histocompatibility complex (MHC) class I pathway in lymphoid malignancies, and transplant setting has been recognised as an important mediator of immune pathogenesis and T cell-mediated immune regulation. However, the precise mechanism of cross-presentation of exogenous antigens in B cells has remained unresolved. Here we have delineated a novel pathway for cross-presentation in B cells, which involves synergistic cooperation of the proteasome and autophagy. After endocytosis, protein antigen is processed through an autophagy- and proteasome-dependent pathway and CD8+ T-cell epitopes are loaded onto MHC class I molecules within the autophagolysomal compartment rather than the conventional secretory pathway, which requires transporters associated with antigen processing-dependent transport. Interestingly, this cross-presentation was critically dependent on valosin-containing protein (VCP)/p97 ATPase through its participation in autophagy. Loss of VCP/p97 ATPase was coincident with accumulation of LC3-II and marked reduction in antigen presentation. These observations provide unique insight on how the autophagy and proteasomal degradation systems interconnect to coordinate MHC class I-restricted cross-presentation in B cells.


Asunto(s)
Autofagia/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Reactividad Cruzada/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/farmacología , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada/efectos de los fármacos , Citomegalovirus/metabolismo , Epítopos de Linfocito T/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Proteína que Contiene Valosina/metabolismo , Proteínas Virales/metabolismo
20.
J Virol ; 89(17): 9137-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26109734

RESUMEN

Here we present evidence for previously unappreciated B-cell immune dysregulation during acute Epstein-Barr virus (EBV)-associated infectious mononucleosis (IM). Longitudinal analyses revealed that patients with acute IM have undetectable EBV-specific neutralizing antibodies and gp350-specific B-cell responses, which were associated with a significant reduction in memory B cells and no evidence of circulating antibody-secreting cells. These observations correlate with dysregulation of tumor necrosis factor family members BAFF and APRIL and increased expression of FAS on circulating B cells.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Herpesvirus Humano 4/inmunología , Mononucleosis Infecciosa/inmunología , Factor Activador de Células B/inmunología , Linfocitos B/virología , Linfocitos T CD8-positivos/inmunología , Proteínas de la Cápside/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/virología , Humanos , Memoria Inmunológica/inmunología , Mononucleosis Infecciosa/virología , Activación de Linfocitos/inmunología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Proteínas de la Matriz Viral/inmunología , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA