Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.496
Filtrar
1.
J Vis Exp ; (209)2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39141526

RESUMO

The identification and characterization of antigen-specific T cells during health and disease remains a key to improving our understanding of immune pathophysiology. The technical challenges of tracking antigen-specific T cell populations within the endogenous T cell repertoire have been greatly advanced by the development of peptide:MHC tetramer reagents. These fluorescently labeled soluble multimers of MHC class I or class II molecules complexed to antigenic peptide epitopes bind directly to T cells with corresponding T cell receptor (TCR) specificity and can, therefore, identify antigen-specific T cell populations in their native state without a requirement for a functional response induced by ex vivo stimulation. For exceedingly rare populations, tetramer-bound T cells can be magnetically enriched to increase the sensitivity and reliability of detection. As the investigation of tissue-resident T cell immunity deepens, there is a pressing need to identify antigen-specific T cells that traffic to and reside in nonlymphoid tissues. In this protocol, we present a detailed set of instructions for the isolation and characterization of antigen-specific T cells present within mouse lungs. This involves the isolation of T cells from digested lung tissue followed by a general T cell magnetic enrichment step and tetramer staining for flow cytometry analysis and sorting. The steps highlighted in this protocol utilize common techniques and readily available reagents, making it accessible for nearly any researcher engaged in mouse T cell immunology, and are highly adaptable for a variety of downstream analyses of any low frequency antigen-specific T cell population residing within the lungs.


Assuntos
Pulmão , Animais , Camundongos , Pulmão/imunologia , Pulmão/citologia , Peptídeos/imunologia , Peptídeos/química , Linfócitos T/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Epitopos de Linfócito T/imunologia
2.
Sci Immunol ; 9(97): eado5295, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38996008

RESUMO

αß T cell receptor (TCR) V(D)J genes code for billions of TCR combinations. However, only some appear on peripheral T cells in any individual because, to mature, thymocytes must react with low affinity but not high affinity with thymus expressed major histocompatibility (MHC)/peptides. MHC proteins are very polymorphic. Different alleles bind different peptides. Therefore, any individual might express many different MHC alleles to ensure that some peptides from an invader are bound to MHC and activate T cells. However, most individuals express limited numbers of MHC alleles. To explore this, we compared the TCR repertoires of naïve CD4 T cells in mice expressing one or two MHC alleles. Unexpectedly, the TCRs in heterozygotes were less diverse that those in the sum of their MHC homozygous relatives. Our results suggest that thymus negative selection cancels out the advantages of increased thymic positive selection in the MHC heterozygotes.


Assuntos
Linfócitos T CD4-Positivos , Heterozigoto , Animais , Camundongos , Linfócitos T CD4-Positivos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/genética , Complexo Principal de Histocompatibilidade/imunologia , Complexo Principal de Histocompatibilidade/genética , Camundongos Endogâmicos C57BL , Timo/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Camundongos Transgênicos
3.
Nat Comput Sci ; 4(7): 510-521, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38987378

RESUMO

T cell receptor (TCR) recognition of foreign peptides presented by major histocompatibility complex protein is a major event in triggering the adaptive immune response to pathogens or cancer. The prediction of TCR-peptide interactions has great importance for therapy of cancer as well as infectious and autoimmune diseases but remains a major challenge, particularly for novel (unseen) peptide epitopes. Here we present TCRen, a structure-based method for ranking candidate unseen epitopes for a given TCR. The first stage of the TCRen pipeline is modeling of the TCR-peptide-major histocompatibility complex structure. Then a TCR-peptide residue contact map is extracted from this structure and used to rank all candidate epitopes on the basis of an interaction score with the target TCR. Scoring is performed using an energy potential derived from the statistics of TCR-peptide contact preferences in existing crystal structures. We show that TCRen has high performance in discriminating cognate versus unrelated peptides and can facilitate the identification of cancer neoepitopes recognized by tumor-infiltrating lymphocytes.


Assuntos
Receptores de Antígenos de Linfócitos T , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/metabolismo , Humanos , Peptídeos/imunologia , Peptídeos/química , Epitopos/imunologia , Epitopos/química , Modelos Moleculares , Neoplasias/imunologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/química , Complexo Principal de Histocompatibilidade/imunologia , Conformação Proteica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo
4.
Front Immunol ; 15: 1436233, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39035001

RESUMO

The peptide presentation by donor and recipient major histocompatibility complex (MHC) molecules is the major driver of T-cell responses in transplantation. In this review, we address an emerging area of interest, the application of immunopeptidome in transplantation, and describe the potential opportunities that exist to use peptides for targeting alloreactive T cells. The immunopeptidome, the set of peptides presented on an individual's MHC, plays a key role in immune surveillance. In transplantation, the immunopeptidome is heavily influenced by MHC-derived peptides, delineating a key subset of the diverse peptide repertoire implicated in alloreactivity. A better understanding of the immunopeptidome in transplantation has the potential to open up new approaches to identify, characterize, longitudinally quantify, and therapeutically target donor-specific T cells and ultimately support more personalized immunotherapies to prevent rejection and promote allograft tolerance.


Assuntos
Rejeição de Enxerto , Peptídeos , Humanos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Peptídeos/imunologia , Animais , Complexo Principal de Histocompatibilidade/imunologia , Transplante de Órgãos , Linfócitos T/imunologia , Apresentação de Antígeno/imunologia
5.
Exp Mol Med ; 56(6): 1461-1471, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38866910

RESUMO

Neoantigens are ideal targets for cancer immunotherapy because they are expressed de novo in tumor tissue but not in healthy tissue and are therefore recognized as foreign by the immune system. Advances in next-generation sequencing and bioinformatics technologies have enabled the quick identification and prediction of tumor-specific neoantigens; however, only a small fraction of predicted neoantigens are immunogenic. To improve the predictability of immunogenic neoantigens, we developed the in silico neoantigen prediction workflows VACINUSpMHC and VACINUSTCR: VACINUSpMHC incorporates physical binding between peptides and MHCs (pMHCs), and VACINUSTCR integrates T cell reactivity to the pMHC complex through deep learning-based pairing with T cell receptors (TCRs) of putative tumor-reactive CD8 tumor-infiltrating lymphocytes (TILs). We then validated our neoantigen prediction workflows both in vitro and in vivo in patients with hepatocellular carcinoma (HCC) and in a B16F10 mouse melanoma model. The predictive abilities of VACINUSpMHC and VACINUSTCR were confirmed in a validation cohort of 8 patients with HCC. Of a total of 118 neoantigen candidates predicted by VACINUSpMHC, 48 peptides were ultimately selected using VACINUSTCR. In vitro validation revealed that among the 48 predicted neoantigen candidates, 13 peptides were immunogenic. Assessment of the antitumor efficacy of the candidate neoepitopes using a VACINUSTCR in vivo mouse model suggested that vaccination with the predicted neoepitopes induced neoantigen-specific T cell responses and enabled the trafficking of neoantigen-specific CD8 + T cell clones into the tumor tissue, leading to tumor suppression. This study showed that the prediction of immunogenic neoantigens can be improved by integrating a tumor-reactive TIL TCR-pMHC ternary complex.


Assuntos
Antígenos de Neoplasias , Linfócitos do Interstício Tumoral , Receptores de Antígenos de Linfócitos T , Antígenos de Neoplasias/imunologia , Animais , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linhagem Celular Tumoral , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Complexo Principal de Histocompatibilidade/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/terapia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/terapia , Linfócitos T CD8-Positivos/imunologia , Feminino , Imunoterapia/métodos
6.
Cancer Lett ; 597: 217062, 2024 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-38878852

RESUMO

Immune checkpoint inhibitors (ICIs) have transformed cancer therapy, yet persistent challenges such as low response rate and significant heterogeneity necessitate attention. The pivotal role of the major histocompatibility complex (MHC) in ICI efficacy, its intricate impacts and potentials as a prognostic marker, warrants comprehensive exploration. This study integrates single-cell RNA sequencing (scRNA-seq), bulk RNA-seq, and spatial transcriptomic analyses to unveil pan-cancer immune characteristics governed by the MHC transcriptional feature (MHC.sig). Developed through scRNA-seq analysis of 663,760 cells across diverse cohorts and validated in 30 solid cancer types, the MHC.sig demonstrates a robust correlation between immune-related genes and infiltrating immune cells, highlighting its potential as a universal pan-cancer marker for anti-tumor immunity. Screening the MHC.sig for therapeutic targets using CRISPR data identifies potential genes for immune therapy synergy and validates its predictive efficacy for ICIs responsiveness across diverse datasets and cancer types. Finally, analysis of cellular communication patterns reveals interactions between C1QC+macrophages and malignant cells, providing insights into potential therapeutic agents and their sensitivity characteristics. This comprehensive analysis positions the MHC.sig as a promising marker for predicting immune therapy outcomes and guiding combinatorial therapeutic strategies.


Assuntos
Complexo Principal de Histocompatibilidade , Neoplasias , Análise de Célula Única , Humanos , Neoplasias/genética , Neoplasias/imunologia , Análise de Célula Única/métodos , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/imunologia , Análise de Sequência de RNA/métodos , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/farmacologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral/imunologia , Microambiente Tumoral/genética , RNA-Seq
7.
Adv Protein Chem Struct Biol ; 140: 327-345, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38762273

RESUMO

Major histocompatibility complex (MHC) tetramers stand as formidable tools within T cell biology, facilitating the exploration and comprehension of immune responses. These artificial molecules, comprising four bound MHC molecules, typically with a specified peptide and a fluorescent label, play a pivotal role in characterizing T cell subsets, monitoring clonal expansion, and unraveling T cell dynamics during responses to infections or immunotherapies. Beyond their applications in T cell biology, MHC tetramers prove valuable in investigating a spectrum of diseases such as infectious diseases, autoimmune disorders, and cancers. Their instrumental role extends to vaccine research and development. Notably, when appropriately configured, tetramers transcend T cell biology research and find utility in exploring natural killer T cells and contributing to specific T cell clonal deletions.


Assuntos
Complexo Principal de Histocompatibilidade , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo
8.
Proc Natl Acad Sci U S A ; 121(22): e2319029121, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38781214

RESUMO

The HapImmuneTM platform exploits covalent inhibitors as haptens for creating major histocompatibility complex (MHC)-presented tumor-specific neoantigens by design, combining targeted therapies with immunotherapy for the treatment of drug-resistant cancers. A HapImmune antibody, R023, recognizes multiple sotorasib-conjugated KRAS(G12C) peptides presented by different human leukocyte antigens (HLAs). This high specificity to sotorasib, coupled with broad HLA-binding capability, enables such antibodies, when reformatted as T cell engagers, to potently and selectively kill sotorasib-resistant KRAS(G12C) cancer cells expressing different HLAs upon sotorasib treatment. The loosening of HLA restriction could increase the patient population that can benefit from this therapeutic approach. To understand the molecular basis for its unconventional binding capability, we used single-particle cryogenic electron microscopy to determine the structures of R023 bound to multiple sotorasib-peptide conjugates presented by different HLAs. R023 forms a pocket for sotorasib between the VH and VL domains, binds HLAs in an unconventional, angled way, with VL making most contacts with them, and makes few contacts with the peptide moieties. This binding mode enables the antibody to accommodate different hapten-peptide conjugates and to adjust its conformation to different HLAs presenting hapten-peptides. Deep mutational scanning validated the structures and revealed distinct levels of mutation tolerance by sotorasib- and HLA-binding residues. Together, our structural information and sequence landscape analysis reveal key features for achieving MHC-restricted recognition of multiple hapten-peptide antigens, which will inform the development of next-generation therapeutic antibodies.


Assuntos
Peptídeos , Humanos , Peptídeos/imunologia , Peptídeos/química , Antígenos HLA/imunologia , Antígenos HLA/metabolismo , Complexo Principal de Histocompatibilidade/imunologia , Haptenos/imunologia , Ligação Proteica , Microscopia Crioeletrônica
9.
Scand J Immunol ; 100(2): e13375, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38750629

RESUMO

Our understanding of the immune response is far from complete, missing out on more detailed explanations that could be provided by molecular insights. To bridge this gap, we introduce the quantum model of T-cell activation. This model suggests that the transfer of energy during protein phosphorylation within T cells is not a continuous flow but occurs in discrete bursts, or 'quanta', of phosphates. This quantized energy transfer is mediated by oscillating cycles of receptor phosphorylation and dephosphorylation, initiated by dynamic 'catch-slip' pulses in the peptide-major histocompatibility complex-T-cell receptor (pMHC-TcR) interactions. T-cell activation is predicated upon achieving a critical threshold of catch-slip pulses at the pMHC-TcR interface. Costimulation is relegated to a secondary role, becoming crucial only when the frequency of pMHC-TcR catch-slip pulses does not meet the necessary threshold for this quanta-based energy transfer. Therefore, our model posits that it is the quantum nature of energy transfer-not the traditional signal I or signal II-that plays the decisive role in T-cell activation. This paradigm shift highlights the importance of understanding T-cell activation through a quantum lens, offering a potentially transformative perspective on immune response regulation.


Assuntos
Ativação Linfocitária , Receptores de Antígenos de Linfócitos T , Linfócitos T , Animais , Humanos , Transferência de Energia , Ativação Linfocitária/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Modelos Imunológicos , Fosforilação , Teoria Quântica , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/imunologia
10.
Pharmacol Res ; 203: 107168, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38583689

RESUMO

Parkinson's disease (PD) is a common neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra and the aggregation of alpha-synuclein (α-syn). The central nervous system (CNS) has previously been considered as an immune-privileged area. However, studies have shown that the immune responses are involved in PD. The major histocompatibility complex (MHC) presents antigens from antigen-presenting cells (APCs) to T lymphocytes, immune responses will be induced. MHCs are expressed in microglia, astrocytes, and dopaminergic neurons. Single nucleotide polymorphisms in MHC are related to the risk of PD. The aggregated α-syn triggers the expression of MHCs by activating glia cells. CD4+ and CD8+ T lymphocytes responses and microglia activation are detected in brains of PD patients. In addiction immune responses further increase blood-brain barrier (BBB) permeability and T cell infiltration in PD. Thus, MHCs are involved in PD through participating in immune and inflammatory responses.


Assuntos
Complexo Principal de Histocompatibilidade , Doença de Parkinson , Animais , Humanos , alfa-Sinucleína/imunologia , alfa-Sinucleína/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Complexo Principal de Histocompatibilidade/imunologia , Microglia/imunologia , Microglia/metabolismo , Doença de Parkinson/imunologia , Doença de Parkinson/genética
11.
Nature ; 618(7967): 1033-1040, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37316667

RESUMO

Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.


Assuntos
Linfócitos T CD4-Positivos , Morte Celular , Imunoterapia , Inflamação , Neoplasias , Microambiente Tumoral , Humanos , Células Apresentadoras de Antígenos/imunologia , Antígeno CD11c/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Morte Celular/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Imunidade Inata , Inflamação/imunologia , Interferons/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/terapia , Microambiente Tumoral/imunologia , Imunoterapia/métodos , Células Matadoras Naturais/imunologia , Células Mieloides/imunologia , Células Th1/citologia , Células Th1/imunologia
12.
Mol Immunol ; 142: 105-119, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34973498

RESUMO

In the late 1980s and early 1990s, the hunt for a transporter molecule ostensibly responsible for the translocation of peptides across the endoplasmic reticulum (ER) membrane yielded the successful discovery of transporter associated with antigen processing (TAP) protein. TAP is a heterodimer complex comprised of TAP1 and TAP2, which utilizes ATP to transport cytosolic peptides into the ER across its membrane. In the ER, together with other components it forms the peptide loading complex (PLC), which directs loading of high affinity peptides onto nascent major histocompatibility complex class I (MHC-I) molecules that are then transported to the cell surface for presentation to CD8+ T cells. TAP also plays a crucial role in transporting peptides into phagosomes and endosomes during cross-presentation in dendritic cells (DCs). Because of the critical role that TAP plays in both classical MHC-I presentation and cross-presentation, its expression and function are often compromised by numerous types of cancers and viruses to evade recognition by cytotoxic CD8 T cells. Here we review the discovery and function of TAP with a major focus on its role in cross-presentation in DCs. We discuss a recently described emergency route of noncanonical cross-presentation that is mobilized in DCs upon TAP blockade to restore CD8 T cell cross-priming. We also discuss the various strategies employed by cancer cells and viruses to target TAP expression or function to evade immunosurveillance - along with some strategies by which the repertoire of peptides presented by cells which downregulate TAP can be targeted as a therapeutic strategy to mobilize a TAP-independent CD8 T cell response. Lastly, we discuss TAP polymorphisms and the role of TAP in inherited disorders.


Assuntos
Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Apresentação de Antígeno/imunologia , Apresentação Cruzada/imunologia , Evasão Tumoral/imunologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/genética , Células Dendríticas/imunologia , Retículo Endoplasmático/metabolismo , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Neoplasias/imunologia , Transporte Proteico/genética , Linfócitos T Citotóxicos/imunologia , Vírus/imunologia
13.
J Biol Chem ; 298(2): 101542, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34968463

RESUMO

The monomorphic antigen-presenting molecule major histocompatibility complex-I-related protein 1 (MR1) presents small-molecule metabolites to mucosal-associated invariant T (MAIT) cells. The MR1-MAIT cell axis has been implicated in a variety of infectious and noncommunicable diseases, and recent studies have begun to develop an understanding of the molecular mechanisms underlying this specialized antigen presentation pathway. However, proteins regulating MR1 folding, loading, stability, and surface expression remain to be identified. Here, we performed a gene trap screen to discover novel modulators of MR1 surface expression through insertional mutagenesis of an MR1-overexpressing clone derived from the near-haploid human cell line HAP1 (HAP1.MR1). The most significant positive regulators identified included ß2-microglobulin, a known regulator of MR1 surface expression, and ATP13A1, a P5-type ATPase in the endoplasmic reticulum (ER) not previously known to be associated with MR1-mediated antigen presentation. CRISPR/Cas9-mediated knockout of ATP13A1 in both HAP1.MR1 and THP-1 cell lines revealed a profound reduction in MR1 protein levels and a concomitant functional defect specific to MR1-mediated antigen presentation. Collectively, these data are consistent with the ER-resident ATP13A1 being a key posttranscriptional determinant of MR1 surface expression.


Assuntos
Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I , Complexo Principal de Histocompatibilidade , Antígenos de Histocompatibilidade Menor , ATPases do Tipo-P , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Antígenos de Histocompatibilidade Menor/imunologia , ATPases do Tipo-P/imunologia
14.
Sci Rep ; 11(1): 19862, 2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34615944

RESUMO

Individual variation in the age of pubertal onset is linked to physical and mental health, yet the factors underlying this variation are poorly understood. Life history theory predicts that individuals at higher risk of mortality due to extrinsic causes such as infectious disease should sexually mature and reproduce earlier, whereas those at lower risk can delay puberty and continue to invest resources in somatic growth. We examined relationships between a genetic predictor of infectious disease resistance, heterozygosity of the major histocompatibility complex (MHC), referred to as the human leukocyte antigen (HLA) gene in humans, and self-reported pubertal timing. In a combined sample of men from Canada (n = 137) and the United States (n = 43), MHC heterozygosity predicted later self-reported pubertal development. These findings suggest a genetic trade-off between immunocompetence and sexual maturation in human males.


Assuntos
Alelos , Heterozigoto , Complexo Principal de Histocompatibilidade/genética , Puberdade/genética , Adolescente , Adulto , Resistência à Doença/genética , Suscetibilidade a Doenças/imunologia , Predisposição Genética para Doença , Humanos , Complexo Principal de Histocompatibilidade/imunologia , Masculino , Puberdade/imunologia , Autorrelato , Adulto Jovem
15.
Front Immunol ; 12: 742881, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34650566

RESUMO

Despite the high number of individuals infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) who develop coronavirus disease 2019 (COVID-19) symptoms worldwide, many exposed individuals remain asymptomatic and/or uninfected and seronegative. This could be explained by a combination of environmental (exposure), immunological (previous infection), epigenetic, and genetic factors. Aiming to identify genetic factors involved in immune response in symptomatic COVID-19 as compared to asymptomatic exposed individuals, we analyzed 83 Brazilian couples where one individual was infected and symptomatic while the partner remained asymptomatic and serum-negative for at least 6 months despite sharing the same bedroom during the infection. We refer to these as "discordant couples". We performed whole-exome sequencing followed by a state-of-the-art method to call genotypes and haplotypes across the highly polymorphic major histocompatibility complex (MHC) region. The discordant partners had comparable ages and genetic ancestry, but women were overrepresented (65%) in the asymptomatic group. In the antigen-presentation pathway, we observed an association between HLA-DRB1 alleles encoding Lys at residue 71 (mostly DRB1*03:01 and DRB1*04:01) and DOB*01:02 with symptomatic infections and HLA-A alleles encoding 144Q/151R with asymptomatic seronegative women. Among the genes related to immune modulation, we detected variants in MICA and MICB associated with symptomatic infections. These variants are related to higher expression of soluble MICA and low expression of MICB. Thus, quantitative differences in these molecules that modulate natural killer (NK) activity could contribute to susceptibility to COVID-19 by downregulating NK cell cytotoxic activity in infected individuals but not in the asymptomatic partners.


Assuntos
Infecções Assintomáticas , COVID-19 , Antígenos de Histocompatibilidade , Complexo Principal de Histocompatibilidade , SARS-CoV-2 , Adulto , Idoso , Brasil , COVID-19/genética , COVID-19/imunologia , Feminino , Predisposição Genética para Doença , Genótipo , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/imunologia , Humanos , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/imunologia , Masculino , Pessoa de Meia-Idade , Sequenciamento do Exoma
16.
Sci Rep ; 11(1): 19056, 2021 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-34561502

RESUMO

Hairy cell leukaemia (HCL) is a rare CD20+ B cell malignancy characterised by rare "hairy" B cells and extensive bone marrow (BM) infiltration. Frontline treatment with the purine analogue cladribine (CDA) results in a highly variable response duration. We hypothesised that analysis of the BM tumour microenvironment would identify prognostic biomarkers of response to CDA. HCL BM immunology pre and post CDA treatment and healthy controls were analysed using Digital Spatial Profiling to assess the expression of 57 proteins using an immunology panel. A bioinformatics pipeline was developed to accommodate the more complex experimental design of a spatially resolved study. Treatment with CDA was associated with the reduction in expression of HCL tumour markers (CD20, CD11c) and increased expression of myeloid markers (CD14, CD68, CD66b, ARG1). Expression of HLA-DR, STING, CTLA4, VISTA, OX40L were dysregulated pre- and post-CDA. Duration of response to treatment was associated with greater reduction in tumour burden and infiltration by CD8 T cells into the BM post-CDA. This is the first study to provide a high multiplex analysis of HCL BM microenvironment demonstrating significant immune dysregulation and identify biomarkers of response to CDA. With validation in future studies, prospective application of these biomarkers could allow early identification and increased monitoring in patients at increased relapse risk post CDA.


Assuntos
Biomarcadores Tumorais/metabolismo , Leucemia de Células Pilosas/patologia , Microambiente Tumoral , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Leucemia de Células Pilosas/imunologia , Leucemia de Células Pilosas/metabolismo , Complexo Principal de Histocompatibilidade/imunologia , Masculino , Pessoa de Meia-Idade , Prognóstico
17.
Sci Rep ; 11(1): 17234, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34446788

RESUMO

Over the past two decades, there has been a great interest in the study of HLA-E-restricted αß T cells during bacterial and viral infections, including recently SARS-CoV-2 infection. Phenotyping of these specific HLA-E-restricted T cells requires new tools such as tetramers for rapid cell staining or sorting, as well as for the identification of new peptides capable to bind to the HLA-E pocket. To this aim, we have developed an optimal photosensitive peptide to generate stable HLA-E/pUV complexes allowing high-throughput production of new HLA-E/peptide complexes by peptide exchange. We characterized the UV exchange by ELISA and improved the peptide exchange readout using size exclusion chromatography. This novel approach for complex quantification is indeed very important to perform tetramerization of MHC/peptide complexes with the high quality required for detection of specific T cells. Our approach allows the rapid screening of peptides capable of binding to the non-classical human HLA-E allele, paving the way for the development of new therapeutic approaches based on the detection of HLA-E-restricted T cells.


Assuntos
Epitopos de Linfócito T/química , Antígenos de Histocompatibilidade Classe I/química , Complexo Principal de Histocompatibilidade/imunologia , Peptídeos/química , Sequência de Aminoácidos , Epitopos de Linfócito T/imunologia , Ensaios de Triagem em Larga Escala , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Técnicas Imunológicas , Processos Fotoquímicos , Ligação Proteica , Conformação Proteica , Linfócitos T Citotóxicos/imunologia , Antígenos HLA-E
18.
Mol Immunol ; 139: 76-86, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34455212

RESUMO

The activation of T cells is triggered by the interactions of T cell receptors (TCRs) with their epitopes, which are peptides presented by major histocompatibility complex (MHC) on the surfaces of antigen presenting cells (APC). While each TCR can only recognize a specific subset from a large repertoire of peptide-MHC (pMHC) complexes, it is very often that peptides in this subset share little sequence similarity. This is known as the specificity and cross-reactivity of T cells, respectively. The binding affinities between different types of TCRs and pMHC are the major driving force to shape this specificity and cross-reactivity in T cell recognition. The binding affinities, furthermore, are determined by the sequence and structural properties at the interfaces between TCRs and pMHC. Fortunately, a wealth of data on binding and structures of TCR-pMHC interactions becomes publicly accessible in online resources, which offers us the opportunity to develop a random forest classifier for predicting the binding affinities between TCR and pMHC based on the structure of their complexes. Specifically, the structure and sequence of a given complex were projected onto a high-dimensional feature space as the input of the classifier, which was then trained by a large-scale benchmark dataset. Based on the cross-validation results, we found that our machine learning model can predict if the binding affinity of a given TCR-pMHC complex is stronger or weaker than a predefined threshold with an overall accuracy approximately around 75 %. The significance of our prediction was estimated by statistical analysis. Moreover, more than 60 % of binding affinities in the ATLAS database can be successfully classified into groups within the range of 2 kcal/mol. Additionally, we show that TCR-pMHC complexes with strong binding affinity prefer hydrophobic interactions between amino acids with large aromatic rings instead of electrostatic interactions. Our results therefore provide insights to design engineered TCRs which enhance the specificity for their targeted epitopes. Taken together, this method can serve as a useful addition to a suite of existing approaches which study binding between TCR and pMHC.


Assuntos
Ativação Linfocitária/imunologia , Aprendizado de Máquina , Complexo Principal de Histocompatibilidade/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Humanos , Ligação Proteica
19.
Nat Methods ; 18(8): 881-892, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34282327

RESUMO

T cells express T cell receptors (TCRs) composed of somatically recombined TCRα and TCRß chains, which mediate recognition of major histocompatibility complex (MHC)-antigen complexes and drive the antigen-specific adaptive immune response to pathogens and cancer. The TCR repertoire in each individual is highly diverse, which allows for recognition of a wide array of foreign antigens, but also presents a challenge in analyzing this response using conventional methods. Recent studies have developed high-throughput sequencing technologies to identify TCR sequences, analyze their antigen specificities using experimental and computational tools, and pair TCRs with transcriptional and epigenetic cell state phenotypes in single cells. In this Review, we highlight these technological advances and describe how they have been applied to discover fundamental insights into T cell-mediated immunity.


Assuntos
Antígenos/imunologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Complexo Principal de Histocompatibilidade/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Análise de Célula Única/métodos , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Humanos
20.
Front Immunol ; 12: 664624, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33995395

RESUMO

Melanized focal changes in white skeletal muscle of farmed Atlantic salmon, "black spots", is a quality problem affecting on average 20% of slaughtered fish. The spots appear initially as "red spots" characterized by hemorrhages and acute inflammation and progress into black spots characterized by chronic inflammation and abundant pigmented cells. Piscine orthoreovirus 1 (PRV-1) was previously found to be associated with macrophages and melano-macrophages in red and black spots. Here we have addressed the inflammatory microenvironment of red and black spots by studying the polarization status of the macrophages and cell mediated immune responses in spots, in both PRV-1 infected and non-infected fish. Samples that had been collected at regular intervals through the seawater production phase in a commercial farm were analyzed by multiplex fluorescent in situ hybridization (FISH) and RT-qPCR methods. Detection of abundant inducible nitric oxide synthase (iNOS2) expressing M1-polarized macrophages in red spots demonstrated a pro-inflammatory microenvironment. There was an almost perfect co-localization with the iNOS2 expression and PRV-1 infection. Black spots, on the other side, had few iNOS2 expressing cells, but a relatively high number of arginase-2 expressing anti-inflammatory M2-polarized macrophages containing melanin. The numerous M2-polarized melano-macrophages in black spots indicate an ongoing healing phase. Co-localization of PRV-1 and cells expressing CD8+ and MHC-I suggests a targeted immune response taking place in the spots. Altogether, this study indicates that PRV-1 induces a pro-inflammatory environment that is important for the pathogenesis of the spots. We do not have indication that infection of PRV-1 is the initial causative agent of this condition.


Assuntos
Microambiente Celular , Doenças dos Peixes/etiologia , Doenças dos Peixes/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Orthoreovirus/fisiologia , Infecções por Reoviridae/veterinária , Salmo salar , Animais , Biomarcadores , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Doenças dos Peixes/patologia , Imunofluorescência , Imuno-Histoquímica , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/patologia , Complexo Principal de Histocompatibilidade/genética , Complexo Principal de Histocompatibilidade/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA