Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Clin Immunol ; 44(6): 147, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38856804

RESUMEN

PURPOSE: Asymptomatic SARS-CoV-2 infections were widely reported during the COVID-19 pandemic, acting as a hidden source of infection. Many existing studies investigating asymptomatic immunity failed to recruit true asymptomatic individuals. Thus, we conducted a longitudinal cohort study to evaluate humoral- and cell-mediated responses to infection and vaccination in well-defined asymptomatic young adults (the Asymptomatic COVID-19 in Education [ACE] cohort). METHODS: Asymptomatic testing services located at three UK universities identified asymptomatic young adults who were subsequently recruited with age- and sex-matched symptomatic and uninfected controls. Blood and saliva samples were collected after SARS-CoV-2 Wuhan infection, and again after vaccination. 51 participant's anti-spike antibody titres, neutralizing antibodies, and spike-specific T-cell responses were measured, against both Wuhan and Omicron B.1.1.529.1. RESULTS: Asymptomatic participants exhibited reduced Wuhan-specific neutralization antibodies pre- and post-vaccination, as well as fewer Omicron-specific neutralization antibodies post-vaccination, compared to symptomatic participants. Lower Wuhan and Omicron-specific IgG titres in asymptomatic individuals were also observed pre- and post-vaccination, compared to symptomatic participants. There were no differences in salivary IgA levels. Conventional flow cytometry analysis and multi-dimensional clustering analysis indicated unvaccinated asymptomatic participants had significantly fewer Wuhan-specific IL-2 secreting CD4+ CD45RA+ T cells and activated CD8+ T cells than symptomatic participants, though these differences dissipated after vaccination. CONCLUSIONS: Asymptomatic infection results in decreased antibody and T cell responses to further exposure to SARS-CoV-2 variants, compared to symptomatic infection. Post-vaccination, antibody responses are still inferior, but T cell immunity increases to match symptomatic subjects, emphasising the importance of vaccination to help protect asymptomatic individuals against future variants.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Infecciones Asintomáticas , COVID-19 , Inmunidad Celular , Inmunidad Humoral , SARS-CoV-2 , Humanos , COVID-19/inmunología , SARS-CoV-2/inmunología , Masculino , Femenino , Anticuerpos Antivirales/sangre , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Adulto Joven , Adulto , Vacunas contra la COVID-19/inmunología , Estudios de Cohortes , Estudios Longitudinales , Vacunación , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Reino Unido/epidemiología , Adolescente , Glicoproteína de la Espiga del Coronavirus/inmunología
2.
Ann Neurol ; 91(1): 89-100, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34687063

RESUMEN

OBJECTIVE: The purpose of this study was to investigate the effect of disease modifying therapies on immune response to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) vaccines in people with multiple sclerosis (MS). METHODS: Four hundred seventy-three people with MS provided one or more dried blood spot samples. Information about coronavirus disease 2019 (COVID-19) and vaccine history, medical, and drug history were extracted from questionnaires and medical records. Dried blood spots were eluted and tested for antibodies to SARS-CoV-2. Antibody titers were partitioned into tertiles with people on no disease modifying therapy as a reference. We calculated the odds ratio of seroconversion (univariate logistic regression) and compared quantitative vaccine response (Kruskal Wallis) following the SARS-CoV-2 vaccine according to disease modifying therapy. We used regression modeling to explore the effect of vaccine timing, treatment duration, age, vaccine type, and lymphocyte count on vaccine response. RESULTS: Compared to no disease modifying therapy, the use of anti-CD20 monoclonal antibodies (odds ratio = 0.03, 95% confidence interval [CI] = 0.01-0.06, p < 0.001) and fingolimod (odds ratio = 0.04; 95% CI = 0.01-0.12) were associated with lower seroconversion following the SARS-CoV-2 vaccine. All other drugs did not differ significantly from the untreated cohort. Both time since last anti-CD20 treatment and total time on treatment were significantly associated with the response to the vaccination. The vaccine type significantly predicted seroconversion, but not in those on anti-CD20 medications. Preliminary data on cellular T-cell immunity showed 40% of seronegative subjects had measurable anti-SARS-CoV-2 T cell responses. INTERPRETATION: Some disease modifying therapies convey risk of attenuated serological response to SARS-CoV-2 vaccination in people with MS. We provide recommendations for the practical management of this patient group. ANN NEUROL 20219999:n/a-n/a.


Asunto(s)
Antirreumáticos/uso terapéutico , Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Huésped Inmunocomprometido , Esclerosis Múltiple/inmunología , Seroconversión/efectos de los fármacos , Adulto , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/tratamiento farmacológico , SARS-CoV-2 , Reino Unido
3.
Immunology ; 165(2): 250-259, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34775604

RESUMEN

Accurate assessment of SARS-CoV-2 immunity is critical in evaluating vaccine efficacy and devising public health policies. Whilst the exact nature of effective immunity remains incompletely defined, SARS-CoV-2-specific T-cell responses are a critical feature that will likely form a key correlate of protection against COVID-19. Here, we developed and optimized a high-throughput whole blood-based assay to determine the T-cell response associated with prior SARS-CoV-2 infection and/or vaccination amongst 231 healthy donors and 68 cancer patients. Following overnight in vitro stimulation with SARS-CoV-2-specific peptides, blood plasma samples were analysed for TH 1-type cytokines. Highly significant differential IFN-γ+ /IL-2+ SARS-CoV-2-specific T-cell responses were seen amongst previously infected COVID-19-positive healthy donors in comparison with unknown / naïve individuals (p < 0·0001). IFN-γ production was more effective at identifying asymptomatic donors, demonstrating higher sensitivity (96·0% vs. 83·3%) but lower specificity (84·4% vs. 92·5%) than measurement of IL-2. A single COVID-19 vaccine dose induced IFN-γ and/or IL-2 SARS-CoV-2-specific T-cell responses in 116 of 128 (90·6%) healthy donors, reducing significantly to 27 of 56 (48·2%) when measured in cancer patients (p < 0·0001). A second dose was sufficient to boost T-cell responses in the majority (90·6%) of cancer patients, albeit IFN-γ+ responses were still significantly lower overall than those induced in healthy donors (p = 0·034). Three-month post-vaccination T-cell responses also declined at a faster rate in cancer patients. Overall, this cost-effective standardizable test ensures accurate and comparable assessments of SARS-CoV-2-specific T-cell responses amenable to widespread population immunity testing, and identifies individuals at greater need of booster vaccinations.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Portador Sano/inmunología , Inmunidad Celular , Inmunogenicidad Vacunal , SARS-CoV-2/inmunología , Células TH1/inmunología , Vacunación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/prevención & control , Femenino , Humanos , Interferón gamma/inmunología , Masculino , Persona de Mediana Edad
4.
Br J Cancer ; 127(9): 1595-1602, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35986086

RESUMEN

BACKGROUND: Recent studies have demonstrated that blocking the PI3Kδ signalling enzyme (by administering a small molecule inhibitor, PI-3065) can potently improve the anti-tumour T-cell response through direct inhibition of Tregs. This treatment also has a negative impact on MDSC numbers but the primary mechanism driving this effect has remained unclear. METHODS: The 4T1 breast cancer mouse model was used in combination with PI-3065 to gain insights into the effect of PI3Kδ inhibition on MDSCs. RESULTS: PI-3065 treatment resulted in a concomitant reduction in MDSC expansion and tumour size. However, targeting Tregs independent of PI-3065 was also associated with reduced tumour volume and MDSC numbers. Surgical removal of tumours resulted in a rapid and significant decline in MDSC numbers, whilst ex vivo studies using cells from PI-3065-treated mice demonstrated no direct effect of the inhibitor on MDSC activity. CONCLUSIONS: Our data suggest that MDSCs are not inhibited directly by PI-3065 treatment but that their reduced recruitment and immunosuppression within the tumour microenvironment is an indirect consequence of PI3Kδ-inhibition-driven tumour control. This indicates that PI3Kδ inhibition drives tumour immunity by breaking down multiple immunosuppressive pathways through both direct mechanisms (on Treg) and indirect mechanisms, secondary to tumour control (on MDSCs).


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias , Animales , Ratones , Linfocitos T Reguladores , Microambiente Tumoral , Proliferación Celular
5.
Eur J Immunol ; 51(2): 331-341, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32920841

RESUMEN

Immune checkpoint inhibitors (antibodies that block the T cell co-inhibitory receptors PD-1/PD-L1 or CTLA-4) have revolutionized the treatment of some forms of cancer. Importantly, combination approaches using drugs that target both pathways have been shown to boost the efficacy of such treatments. Subsequently, several other T cell inhibitory receptors have been identified for the development of novel immune checkpoint inhibitors. Included in this list is the co-inhibitory receptor lymphocyte activation gene-3 (LAG-3), which is upregulated on T cells extracted from tumor sites that have suppressive or exhausted phenotypes. However, the molecular rules that govern the function of LAG-3 are still not understood. Using surface plasmon resonance combined with a novel bead-based assay (AlphaScreenTM ), we demonstrate that LAG-3 can directly and specifically interact with intact human leukocyte antigen class II (HLA-II) heterodimers. Unlike the homologue CD4, which has an immeasurably weak affinity using these biophysical approaches, LAG-3 binds with low micromolar affinity. We further validated the interaction at the cell surface by staining LAG-3+ cells with pHLA-II-multimers. These data provide new insights into the mechanism by which LAG-3 initiates T cell inhibition.


Asunto(s)
Antígenos CD/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Antígenos HLA/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Antígenos CD4/inmunología , Línea Celular Tumoral , Humanos , Inhibidores de Puntos de Control Inmunológico/inmunología , Células Jurkat , Neoplasias/inmunología , Proteína del Gen 3 de Activación de Linfocitos
6.
Nucleic Acids Res ; 48(D1): D1057-D1062, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31588507

RESUMEN

Here, we report an update of the VDJdb database with a substantial increase in the number of T-cell receptor (TCR) sequences and their cognate antigens. The update further provides a new database infrastructure featuring two additional analysis modes that facilitate database querying and real-world data analysis. The increased yield of TCR specificity identification methods and the overall increase in the number of studies in the field has allowed us to expand the database more than 5-fold. Furthermore, several new analysis methods are included. For example, batch annotation of TCR repertoire sequencing samples allows for annotating large datasets on-line. Using recently developed bioinformatic methods for TCR motif mining, we have built a reduced set of high-quality TCR motifs that can be used for both training TCR specificity predictors and matching against TCRs of interest. These additions enhance the versatility of the VDJdb in the task of exploring T-cell antigen specificities. The database is available at https://vdjdb.cdr3.net.


Asunto(s)
Biología Computacional/métodos , Bases de Datos Genéticas , Motivos de Nucleótidos , Receptores de Antígenos de Linfocitos T/genética , Recombinación V(D)J , Secuencia de Aminoácidos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Posición Específica de Matrices de Puntuación , Receptores de Antígenos de Linfocitos T/química , Análisis de Secuencia de ADN , Programas Informáticos , Navegador Web
7.
Immunology ; 163(4): 389-398, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33638871

RESUMEN

Oncolytic viruses possess the ability to infect, replicate and lyse malignantly transformed tumour cells. This oncolytic activity amplifies the therapeutic advantage and induces a form of immunogenic cell death, characterized by increased CD8 + T-cell infiltration into the tumour microenvironment. This important feature of oncolytic viruses can result in the warming up of immunologically 'cold' tumour types, presenting the enticing possibility that oncolytic virus treatment combined with immunotherapies may enhance efficacy. In this review, we assess some of the most promising candidates that might be used for oncolytic virotherapy: immunotherapy combinations. We assess their potential as separate agents or as agents combined into a single therapy, where the immunotherapy is encoded within the genome of the oncolytic virus. The development of such advanced agents will require increasingly sophisticated model systems for their preclinical assessment and evaluation. In vivo rodent model systems are fraught with limitations in this regard. Oncolytic viruses replicate selectively within human cells and therefore require human xenografts in immune-deficient mice for their evaluation. However, the use of immune-deficient rodent models hinders the ability to study immune responses against any immunomodulatory transgenes engineered within the viral genome and expressed within the tumour microenvironment. There has therefore been a shift towards the use of more sophisticated ex vivo patient-derived model systems based on organoids and explant co-cultures with immune cells, which may be more predictive of efficacy than contrived and artificial animal models. We review the best of those model systems here.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/tendencias , Neoplasias/inmunología , Viroterapia Oncolítica/tendencias , Virus Oncolíticos/fisiología , Animales , Linfocitos T CD8-positivos/trasplante , Terapia Combinada , Modelos Animales de Enfermedad , Humanos , Inmunización , Ratones , Neoplasias/terapia , Ratas , Microambiente Tumoral
8.
Br J Cancer ; 124(9): 1552-1555, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33674735

RESUMEN

BACKGROUND: The T cell cytokine profile is a key prognostic indicator of post-surgical outcome for colorectal cancer (CRC). Whilst TH1 (IFN-γ+) cell-mediated responses generated in CRC are well documented and are associated with improved survival, antigen-specific TH17 (IL-17A+) responses have not been similarly measured. METHODS: We sought to determine the cytokine profile of circulating tumour antigen-(5T4/CEA) specific T cells of 34 CRC patients to address whether antigen-specific IL-17A responses were detectable and whether these were distinct to IFN-γ responses. RESULTS: As with IFN-γ-producing T cells, anti-5T4/CEA TH17 responses were detectable predominantly in early stage (TNM I/II) CRC patients. Moreover, whilst IL-17A was always produced in association with IFN-γ, this release was mainly from two distinct T cell populations rather than by 'dual producing' T cells. Patients mounting both tumour-specific TH1+/TH17+ responses exhibited prolonged relapse-free survival. CONCLUSIONS: Tumour antigen-specific TH17 responses play a beneficial role in preventing post-operative colorectal tumour recurrence.


Asunto(s)
Antígenos de Neoplasias/inmunología , Neoplasias Colorrectales/inmunología , Cirugía Colorrectal/mortalidad , Interleucina-17/inmunología , Recurrencia Local de Neoplasia/inmunología , Células TH1/inmunología , Estudios de Casos y Controles , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Femenino , Estudios de Seguimiento , Humanos , Interferón gamma/metabolismo , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Pronóstico , Tasa de Supervivencia
9.
J Biol Chem ; 294(52): 20246-20258, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31619516

RESUMEN

CD4+ T-cells recognize peptide antigens, in the context of human leukocyte antigen (HLA) class II molecules (HLA-II), which through peptide-flanking residues (PFRs) can extend beyond the limits of the HLA binding. The role of the PFRs during antigen recognition is not fully understood; however, recent studies have indicated that these regions can influence T-cell receptor (TCR) affinity and pHLA-II stability. Here, using various biochemical approaches including peptide sensitivity ELISA and ELISpot assays, peptide-binding assays and HLA-II tetramer staining, we focused on CD4+ T-cell responses against a tumor antigen, 5T4 oncofetal trophoblast glycoprotein (5T4), which have been associated with improved control of colorectal cancer. Despite their weak TCR-binding affinity, we found that anti-5T4 CD4+ T-cells are polyfunctional and that their PFRs are essential for TCR recognition of the core bound nonamer. The high-resolution (1.95 Å) crystal structure of HLA-DR1 presenting the immunodominant 20-mer peptide 5T4111-130, combined with molecular dynamic simulations, revealed how PFRs explore the HLA-proximal space to contribute to antigen reactivity. These findings advance our understanding of what constitutes an HLA-II epitope and indicate that PFRs can tune weak affinity TCR-pHLA-II interactions.


Asunto(s)
Epítopos/inmunología , Antígeno HLA-DR1/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Cristalografía por Rayos X , Epítopos/química , Epítopos/metabolismo , Antígeno HLA-DR1/química , Antígeno HLA-DR1/inmunología , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Simulación de Dinámica Molecular , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
10.
Eur J Immunol ; 49(7): 1052-1066, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31091334

RESUMEN

The HLA-A*02:01-restricted decapeptide EAAGIGILTV, derived from melanoma antigen recognized by T-cells-1 (MART-1) protein, represents one of the best-studied tumor associated T-cell epitopes, but clinical results targeting this peptide have been disappointing. This limitation may reflect the dominance of the nonapeptide, AAGIGILTV, at the melanoma cell surface. The decapeptide and nonapeptide are presented in distinct conformations by HLA-A*02:01 and TCRs from clinically relevant T-cell clones recognize the nonapeptide poorly. Here, we studied the MEL5 TCR that potently recognizes the nonapeptide. The structure of the MEL5-HLA-A*02:01-AAGIGILTV complex revealed an induced fit mechanism of antigen recognition involving altered peptide-MHC anchoring. This "flexing" at the TCR-peptide-MHC interface to accommodate the peptide antigen explains previously observed incongruences in this well-studied system and has important implications for future therapeutic approaches. Finally, this study expands upon the mechanisms by which molecular plasticity can influence antigen recognition by T cells.


Asunto(s)
Epítopos Inmunodominantes/metabolismo , Inmunoterapia Adoptiva/métodos , Antígeno MART-1/metabolismo , Melanoma/inmunología , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Aminoácidos , Presentación de Antígeno , Sitios de Unión , Células Cultivadas , Células Clonales , Antígeno HLA-A2/química , Antígeno HLA-A2/metabolismo , Humanos , Activación de Linfocitos , Antígeno MART-1/química , Melanoma/terapia , Péptidos/química , Unión Proteica , Conformación Proteica , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/trasplante
11.
Cancer Immunol Immunother ; 69(10): 2063-2073, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32447412

RESUMEN

Although metastatic disease is responsible for the majority of cancer deaths, tests of novel immunotherapies in mouse tumour models often focus on primary tumours without determining whether these therapies also target metastatic disease. This study examined the impact of depleting Foxp3+ regulatory T cells (Treg), on lung metastases, using a mouse model of breast cancer. After Treg-depletion, generation of an immune response to the primary tumour was a critical determinant for limiting development of metastasis. Indeed, resection of the primary tumour abrogated any effect of Treg-depletion on metastases. In addition, whilst the immune response, generated by the primary tumour, prevented metastases development, it had little impact on controlling established disease. Collectively, the data indicate that metastatic cells in the lung are not controlled by immune responses induced by the primary tumour. These findings indicate that targeting Tregs alone will not suffice for treating lung metastases.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Pulmonares/inmunología , Depleción Linfocítica/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Reguladores/inmunología , Neoplasias de la Mama Triple Negativas/inmunología , Animales , Femenino , Factores de Transcripción Forkhead/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Ratones , Linfocitos T Reguladores/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/terapia
12.
Cancer Immunol Immunother ; 68(2): 247-256, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30406375

RESUMEN

The oncofoetal antigen 5T4 is a promising T cell target in the context of colorectal cancer, as demonstrated by a recent clinical study where 5T4-specific T cell responses, induced by vaccination or cyclophosphamide, were associated with a significantly prolonged survival of patients with metastatic disease. Whilst Th1-type (IFN-γ+) responses specific to 5T4, and other oncofoetal antigens, are often readily detectable in early stage CRC patients and healthy donors, their activity is suppressed as the cancer progresses by CD4+CD25hiFoxp3+ regulatory T cells (Treg) which contribute to the immunosuppressive environment conducive to tumour growth. This study mapped the fine specificity of Th1 and Treg cell responses to the 5T4 protein. Surprisingly, both immunogenic peptides and those recognised by Tregs clustered in the same HLA-DR transcending epitope-rich hotspots within the 5T4 protein. Similarly, regions of low Th1-cell immunogenicity also did not contain peptides capable of stimulating Tregs, further supporting the notion that Treg and Th1 cells recognise the same peptides. Understanding the rules which govern the balance of Th1 and Treg cells responding to a given peptide specificity is, therefore, of fundamental importance to designing strategies for manipulating the balance in favour of Th1 cells, and thus the most effective anti-cancer T cell responses.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias Colorrectales/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T/inmunología , Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Neoplasias Colorrectales/metabolismo , Diseño de Fármacos , Epítopos/inmunología , Epítopos/metabolismo , Antígenos HLA-DR/inmunología , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Péptidos/inmunología , Péptidos/metabolismo , Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo
13.
BMC Gastroenterol ; 19(1): 98, 2019 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221083

RESUMEN

BACKGROUND: The relationship between intestinal epithelial integrity and the development of intestinal disease is of increasing interest. A reduction in mucosal integrity has been associated with ulcerative colitis, Crohn's disease and potentially could have links with colorectal cancer development. The Ussing chamber system can be utilised as a valuable tool for measuring gut integrity. Here we describe step-by-step methodology required to measure intestinal permeability of both mouse and human colonic tissue samples ex vivo, using the latest equipment and software. This system can be modified to accommodate other tissues. METHODS: An Ussing chamber was constructed and adapted to support both mouse and human tissue to measure intestinal permeability, using paracellular flux and electrical measurements. Two mouse models of intestinal inflammation (dextran sodium sulphate treatment and T regulatory cell depletion using C57BL/6-FoxP3DTR mice) were used to validate the system along with human colonic biopsy samples. RESULTS: Distinct regional differences in permeability were consistently identified within mouse and healthy human colon. In particular, mice showed increased permeability in the mid colonic region. In humans the left colon is more permeable than the right. Furthermore, inflammatory conditions induced chemically or due to autoimmunity reduced intestinal integrity, validating the use of the system. CONCLUSIONS: The Ussing chamber has been used for many years to measure barrier function. However, a clear and informative methods paper describing the setup of modern equipment and step-by-step procedure to measure mouse and human intestinal permeability isn't available. The Ussing chamber system methodology we describe provides such detail to guide investigation of gut integrity.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Electrodiagnóstico/instrumentación , Mucosa Intestinal/metabolismo , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Electrodiagnóstico/métodos , Fluorescencia , Humanos , Ratones , Ratones Endogámicos C57BL , Permeabilidad
14.
Proc Natl Acad Sci U S A ; 113(20): 5682-7, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27114505

RESUMEN

The αß T-cell coreceptor CD4 enhances immune responses more than 1 million-fold in some assays, and yet the affinity of CD4 for its ligand, peptide-major histocompatibility class II (pMHC II) on antigen-presenting cells, is so weak that it was previously unquantifiable. Here, we report that a soluble form of CD4 failed to bind detectably to pMHC II in surface plasmon resonance-based assays, establishing a new upper limit for the solution affinity at 2.5 mM. However, when presented multivalently on magnetic beads, soluble CD4 bound pMHC II-expressing B cells, confirming that it is active and allowing mapping of the native coreceptor binding site on pMHC II. Whereas binding was undetectable in solution, the affinity of the CD4/pMHC II interaction could be measured in 2D using CD4- and adhesion molecule-functionalized, supported lipid bilayers, yielding a 2D Kd of ∼5,000 molecules/µm(2) This value is two to three orders of magnitude higher than previously measured 2D Kd values for interacting leukocyte surface proteins. Calculations indicated, however, that CD4/pMHC II binding would increase rates of T-cell receptor (TCR) complex phosphorylation by threefold via the recruitment of Lck, with only a small, 2-20% increase in the effective affinity of the TCR for pMHC II. The affinity of CD4/pMHC II therefore seems to be set at a value that increases T-cell sensitivity by enhancing phosphorylation, without compromising ligand discrimination.


Asunto(s)
Antígenos CD4/química , Antígeno HLA-A24/química , Cadenas HLA-DRB1/química , Sitios de Unión , Antígenos CD4/metabolismo , Células HEK293 , Antígeno HLA-A24/metabolismo , Cadenas HLA-DRB1/metabolismo , Humanos , Proteínas de Unión a Maltosa/química , Modelos Moleculares , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Resonancia por Plasmón de Superficie
15.
Immunology ; 154(1): 62-68, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29460448

RESUMEN

The power of T cells for cancer treatment has been demonstrated by the success of co-inhibitory receptor blockade and adoptive T-cell immunotherapies. These treatments are highly successful for certain cancers, but are often personalized, expensive and associated with harmful side effects. Other T-cell-modulating drugs may provide additional means of improving immune responses to tumours without these disadvantages. Conventional chemotherapeutic drugs are traditionally used to target cancers directly; however, it is clear that some also have significant immune-modulating effects that can be harnessed to target tumours. Cyclophosphamide is one such drug; used at lower doses than in mainstream chemotherapy, it can perturb immune homeostasis, tipping the balance towards generation of anti-tumour T-cell responses and control of cancer growth. This review discusses its growing reputation as an immune-modulator whose multiple effects synergize with the microbiota to tip the balance towards tumour immunity offering widespread benefits as a safe, and relatively inexpensive component of cancer immunotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Ciclofosfamida/uso terapéutico , Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Linfocitos T/efectos de los fármacos , Animales , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Neoplasias/patología , Linfocitos T/inmunología , Resultado del Tratamiento , Escape del Tumor/efectos de los fármacos
16.
PLoS Pathog ; 12(12): e1006050, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27926930

RESUMEN

CD4+ T cells support host defence against herpesviruses and other viral pathogens. We identified that CD4+ T cells from systemic and mucosal tissues of hosts infected with the ß-herpesviridae human cytomegalovirus (HCMV) or murine cytomegalovirus (MCMV) express the regulatory cytokine interleukin (IL)-10. IL-10+CD4+ T cells co-expressed TH1-associated transcription factors and chemokine receptors. Mice lacking T cell-derived IL-10 elicited enhanced antiviral T cell responses and restricted MCMV persistence in salivary glands and secretion in saliva. Thus, IL-10+CD4+ T cells suppress antiviral immune responses against CMV. Expansion of this T-cell population in the periphery was promoted by IL-27 whereas mucosal IL-10+ T cell responses were ICOS-dependent. Infected Il27rα-deficient mice with reduced peripheral IL-10+CD4+ T cell accumulation displayed robust T cell responses and restricted MCMV persistence and shedding. Temporal inhibition experiments revealed that IL-27R signaling during initial infection was required for the suppression of T cell immunity and control of virus shedding during MCMV persistence. IL-27 production was promoted by type-I IFN, suggesting that ß-herpesviridae exploit the immune-regulatory properties of this antiviral pathway to establish chronicity. Further, our data reveal that cytokine signaling events during initial infection profoundly influence virus chronicity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Interferón Tipo I/inmunología , Interleucina-27/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Interleucina-10/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Immunology ; 150(4): 389-396, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28009488

RESUMEN

Eukaryotic forms of life have been continually invaded by microbes and larger multicellular parasites, such as helminths. Over a billion years ago bacterial endosymbionts permanently colonized eukaryotic cells leading to recognized organelles with a distinct genetic lineage, such as mitochondria and chloroplasts. Colonization of our skin and mucosal surfaces with bacterial commensals is now known to be important for host health. However, the contribution of chronic virus and parasitic infections to immune homeostasis is being increasingly questioned. Persistent infection does not necessarily equate to exhibiting a chronic illness: healthy hosts (e.g. humans) have chronic viral and parasitic infections with no evidence of disease. Indeed, there are now examples of complex interactions between these microbes and hosts that seem to confer an advantage to the host at a particular time, suggesting that the relationship has progressed along an axis from parasitic to commensal to one of a mutualistic symbiosis. This concept is explored using examples from viruses and parasites, considering how the relationships may be not only detrimental but also beneficial to the human host.


Asunto(s)
Enfermedades Transmisibles/inmunología , Interacciones Huésped-Parásitos , Inmunidad Mucosa , Enfermedades Parasitarias/inmunología , Virosis/inmunología , Animales , Enfermedad Crónica , Homeostasis , Humanos , Simbiosis
19.
J Immunol ; 195(12): 5827-36, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26553072

RESUMEN

Fluorochrome-conjugated peptide-MHC (pMHC) class I multimers are staple components of the immunologist's toolbox, enabling reliable quantification and analysis of Ag-specific CD8(+) T cells irrespective of functional outputs. In contrast, widespread use of the equivalent pMHC class II (pMHC-II) reagents has been hindered by intrinsically weaker TCR affinities for pMHC-II, a lack of cooperative binding between the TCR and CD4 coreceptor, and a low frequency of Ag-specific CD4(+) T cell populations in the peripheral blood. In this study, we show that peptide flanking regions, extending beyond the central nonamer core of MHC-II-bound peptides, can enhance TCR-pMHC-II binding and T cell activation without loss of specificity. Consistent with these findings, pMHC-II multimers incorporating peptide flanking residue modifications proved superior for the ex vivo detection, characterization, and manipulation of Ag-specific CD4(+) T cells, highlighting an unappreciated feature of TCR-pMHC-II interactions.


Asunto(s)
Antígenos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Separación Celular/métodos , Antígenos HLA/inmunología , Fragmentos de Péptidos/metabolismo , Línea Celular , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Activación de Linfocitos , Unión Proteica , Receptores de Antígenos de Linfocitos T/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T
20.
J Immunol ; 194(1): 463-74, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452566

RESUMEN

Fluorochrome-conjugated peptide-MHC (pMHC) multimers are commonly used in combination with flow cytometry for direct ex vivo visualization and characterization of Ag-specific T cells, but these reagents can fail to stain cells when TCR affinity and/or TCR cell-surface density are low. pMHC multimer staining of tumor-specific, autoimmune, or MHC class II-restricted T cells can be particularly challenging, as these T cells tend to express relatively low-affinity TCRs. In this study, we attempted to improve staining using anti-fluorochrome unconjugated primary Abs followed by secondary staining with anti-Ab fluorochrome-conjugated Abs to amplify fluorescence intensity. Unexpectedly, we found that the simple addition of an anti-fluorochrome unconjugated Ab during staining resulted in considerably improved fluorescence intensity with both pMHC tetramers and dextramers and with PE-, allophycocyanin-, or FITC-based reagents. Importantly, when combined with protein kinase inhibitor treatment, Ab stabilization allowed pMHC tetramer staining of T cells even when the cognate TCR-pMHC affinity was extremely low (KD >1 mM) and produced the best results that we have observed to date. We find that this inexpensive addition to pMHC multimer staining protocols also allows improved recovery of cells that have recently been exposed to Ag, improvements in the recovery of self-specific T cells from PBMCs or whole-blood samples, and the use of less reagent during staining. In summary, Ab stabilization of pMHC multimers during T cell staining extends the range of TCR affinities that can be detected, yields considerably enhanced staining intensities, and is compatible with using reduced amounts of these expensive reagents.


Asunto(s)
Citometría de Flujo/métodos , Técnica del Anticuerpo Fluorescente/métodos , Receptores de Antígenos de Linfocitos T/inmunología , Coloración y Etiquetado/métodos , Linfocitos T/inmunología , Anticuerpos/química , Anticuerpos/inmunología , Células Cultivadas , Colorantes Fluorescentes/química , Humanos , Ficocianina/química , Unión Proteica/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA