Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 144
Filter
1.
Transplant Proc ; 56(8): 1841-1846, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232921

ABSTRACT

BACKGROUND: Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) with TCR αß+/CD19+ cell depletion is a promising therapeutic alternative for children with nonmalignant hematologic disorders, especially in low-income countries where finding a compatible donor is challenging. The use of this transplantation approach for nonmalignant hematologic disorders has not been previously described in the Peruvian pediatric population. METHODS: We present the outcomes of children under 19 with nonmalignant hematologic disorders who underwent haplo-HSCT with TCR αß+/CD19+ cell depletion between 2018-2022 at a referral center in Lima, Peru. Survival probabilities and cumulative incidence functions were calculated using the Kaplan-Meier method. RESULTS: A total of 17 children aged between 1 to 18.6 years (median = 9.7 years) were included. The follow-up period ranged from 10 days to 66.20 months, with a median of 4.34 months. The probability of overall survival, event-free survival, and failure-free survival was 33.70%, 31.40%, and 68.8%, respectively. The incidence rate of graft failure was 49.80%, while the mortality rate not associated with graft failure was 18.8%. The incidence rate of acute graft-versus-host disease (GvHD) was 25.60%, and the incidence rate of viral infections was 59.40%. CONCLUSIONS: The high incidence rates of graft failure and viral infections suggest that these factors may negatively impact the survival of children with nonmalignant hematologic disorders who undergo haplo-HSCT with TCR αß+/CD19+ cell depletion. Therefore, optimizing the current conditioning regimens and ensuring timely access to first, second, and third-line antivirals is crucial to improve the survival of these patients.


Subject(s)
Antigens, CD19 , Graft vs Host Disease , Hematologic Diseases , Hematopoietic Stem Cell Transplantation , Transplantation, Haploidentical , Humans , Child , Adolescent , Child, Preschool , Hematopoietic Stem Cell Transplantation/adverse effects , Peru , Male , Female , Infant , Hematologic Diseases/therapy , Hematologic Diseases/surgery , Graft vs Host Disease/immunology , Receptors, Antigen, T-Cell, alpha-beta , Treatment Outcome , Lymphocyte Depletion , Retrospective Studies
2.
J Neuroinflammation ; 20(1): 135, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37264394

ABSTRACT

INTRODUCTION: Gut microbiota plays a critical role in the regulation of immune homeostasis. Accordingly, several autoimmune disorders have been associated with dysbiosis in the gut microbiota. Notably, the dysbiosis associated with central nervous system (CNS) autoimmunity involves a substantial reduction of bacteria belonging to Clostridia clusters IV and XIVa, which constitute major producers of short-chain fatty acids (SCFAs). Here we addressed the role of the surface receptor-mediated effects of SCFAs on mucosal T-cells in the development of CNS autoimmunity. METHODS: To induce CNS autoimmunity, we used the mouse model of experimental autoimmune encephalomyelitis (EAE) induced by immunization with the myelin oligodendrocyte glycoprotein (MOG)-derived peptide (MOG35-55 peptide). To address the effects of GPR43 stimulation on colonic TCRαß+ T-cells upon CNS autoimmunity, mucosal lymphocytes were isolated and stimulated with a selective GPR43 agonist ex vivo and then transferred into congenic mice undergoing EAE. Several subsets of lymphocytes infiltrating the CNS or those present in the gut epithelium and gut lamina propria were analysed by flow cytometry. In vitro migration assays were conducted with mucosal T-cells using transwells. RESULTS: Our results show a sharp and selective reduction of intestinal propionate at the peak of EAE development, accompanied by increased IFN-γ and decreased IL-22 in the colonic mucosa. Further analyses indicated that GPR43 was the primary SCFAs receptor expressed on T-cells, which was downregulated on colonic TCRαß+ T-cells upon CNS autoimmunity. The pharmacologic stimulation of GPR43 increased the anti-inflammatory function and reduced the pro-inflammatory features in several TCRαß+ T-cell subsets in the colonic mucosa upon EAE development. Furthermore, GPR43 stimulation induced the arrest of CNS-autoreactive T-cells in the colonic lamina propria, thus avoiding their infiltration into the CNS and dampening the disease development. Mechanistic analyses revealed that GPR43-stimulation on mucosal TCRαß+ T-cells inhibits their CXCR3-mediated migration towards CXCL11, which is released from the CNS upon neuroinflammation. CONCLUSIONS: These findings provide a novel mechanism involved in the gut-brain axis by which bacterial-derived products secreted in the gut mucosa might control the CNS tropism of autoreactive T-cells. Moreover, this study shows GPR43 expressed on T-cells as a promising therapeutic target for CNS autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Receptors, Antigen, T-Cell, alpha-beta , Mice , Animals , Autoimmunity , Dysbiosis , Central Nervous System , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptides , Mice, Inbred C57BL
3.
J Immunol ; 209(5): 938-949, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35948399

ABSTRACT

RAG1/RAG2 (RAG) endonuclease-mediated assembly of diverse lymphocyte Ag receptor genes by V(D)J recombination is critical for the development and immune function of T and B cells. The RAG1 protein contains a ubiquitin ligase domain that stabilizes RAG1 and stimulates RAG endonuclease activity in vitro. We report in this study that mice with a mutation that inactivates the Rag1 ubiquitin ligase in vitro exhibit decreased rearrangements and altered repertoires of TCRß and TCRα genes in thymocytes and impaired thymocyte developmental transitions that require the assembly and selection of functional TCRß and/or TCRα genes. These Rag1 mutant mice present diminished positive selection and superantigen-mediated negative selection of conventional αß T cells, decreased genesis of invariant NK T lineage αß T cells, and mature CD4+ αß T cells with elevated autoimmune potential. Our findings reveal that the Rag1 ubiquitin ligase domain functions in vivo to stimulate TCRß and TCRα gene recombination and influence differentiation of αß T lineage cells, thereby establishing replete diversity of αß TCRs and populations of αß T cells while restraining generation of potentially autoreactive conventional αß T cells.


Subject(s)
Homeodomain Proteins , Receptors, Antigen, T-Cell, alpha-beta , Ubiquitin , Animals , Cell Lineage , Endonucleases/genetics , Homeodomain Proteins/genetics , Ligases/genetics , Mice , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Superantigens , V(D)J Recombination/genetics
4.
Mol Immunol ; 144: 35-43, 2022 04.
Article in English | MEDLINE | ID: mdl-35176559

ABSTRACT

In mammals, the T lymphocyte receptor (TCR) is a multiprotein complex formed by the proteins TCRα, TCRß, CD3ε, CD3γ, CD3δ, and CD3ζ. It is responsible for recognizing antigens processed and presented by antigen-presenting cells (APC). The TCR is located at the cytoplasmic membrane of the T lymphocyte but is functional assembled in the rough endoplasmic reticulum (RER). Most of the available information on TCR constituents in salmonids comes from numerous nucleotide sequences available in different databases. In this work, by in silico homology modeling, we generated the TCRαß/CD3 complex of rainbow trout (Oncorhynchus mykiss) and characterized the structure of the different proteins and their potential interactions. The results show that the main structural features described in mammalian TCR/CD3 are present in the model predicted for trout. Furthermore, we highlighted several aminoacidic interactions between TCRα, TCRß, CD3γδ, and CD3ε. In silico structural analyses suggest that trout TCRαß complex would fit similarly to that described for mammals. Herein, we explore the implications of the modeled trout complex and the leukocyte phenotypes, mainly associated with different regulation mechanisms of trout TCRαß/CD3 subunits gene expression or may be due to differences in the assembly process of the complex in the RER. However, further studies will be needed to study deeper the mechanisms involved.


Subject(s)
Oncorhynchus mykiss , Receptors, Antigen, T-Cell, alpha-beta , Animals , CD3 Complex , Mammals , Receptor-CD3 Complex, Antigen, T-Cell , Receptors, Antigen, T-Cell/metabolism
5.
Blood Adv ; 6(4): 1175-1185, 2022 02 22.
Article in English | MEDLINE | ID: mdl-34872106

ABSTRACT

Unrelated donor (URD) hematopoietic stem cell transplant (HSCT) is associated with an increased risk of severe graft-versus-host disease (GVHD). TCRαß/CD19 depletion may reduce this risk, whereas maintaining graft-versus-leukemia. Outcome data with TCRαß/CD19 depletion generally describe haploidentical donors, with relatively few URDs. We hypothesized that TCRαß/CD19-depletion would attenuate the risks of GVHD and relapse for URD HSCT. Sixty pediatric and young adult (YA) patients with hematologic malignancies who lacked a matched-related donor were enrolled at 2 large pediatric transplantation centers between October 2014 and September 2019. All patients with acute leukemia had minimal residual disease testing, and DP typing was available for 77%. All patients received myeloablative total body irradiation- or busulfan-based conditioning with no posttransplant immune suppression. Engraftment occurred in 98%. Four-year overall survival was 69% (95% confidence interval [CI], 52%-81%), and leukemia-free survival was 64% (95% CI, 48%-76%), with no difference between lymphoid and myeloid malignancies (P = .6297 and P = .5441, respectively). One patient (1.7%) experienced primary graft failure. Relapse occurred in 11 patients (3-year cumulative incidence, 21%; 95% CI, 11-34), and 8 patients (cumulative incidence, 15%; 95% CI, 6.7-26) experienced nonrelapse mortality. Grade III to IV acute GVHD was seen in 8 patients (13%), and 14 patients (26%) developed chronic GVHD, of which 6 (11%) had extensive disease. Nonpermissive DP mismatch was associated with higher likelihood of acute GVHD (odds ratio, 16.50; 95% CI, 1.67-163.42; P = .0166) but not with the development of chronic GVHD. URD TCRαß/CD19-depleted peripheral HSCT is a safe and effective approach to transplantation for children/YAs with leukemia. This trial was registered at www.clinicaltrials.gov as #NCT02323867.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Acute Disease , Antigens, CD19 , Child , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukemia, Myeloid, Acute/therapy , Receptors, Antigen, T-Cell, alpha-beta , Recurrence , T-Lymphocytes , Unrelated Donors , Young Adult
6.
Immunology ; 165(4): 371-385, 2022 04.
Article in English | MEDLINE | ID: mdl-34939192

ABSTRACT

Double-negative (DN) T cells are present at relatively low frequencies in human peripheral blood, and are characterized as expressing the alpha-beta or gamma-delta T-cell receptor (TCR), but not the CD4 nor the CD8 co-receptors. Despite their low frequencies, these cells are potent producers of cytokines and, thus, are key orchestrators of immune responses. DN T cells were initially associated with induction of peripheral immunological tolerance and immunomodulatory activities related to disease prevention. However, other studies demonstrated that these cells can also display effector functions associated with pathology development. This apparent contradiction highlighted the heterogeneity of the DN T-cell population. Here, we review phenotypic and functional characteristics of DN T cells, emphasizing their role in human diseases. The need for developing biomarkers to facilitate the translation of studies from animal models to humans will also be discussed. Finally, we will examine DN T cells as promising therapeutic targets to prevent or inhibit human disease development.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocyte Subsets , Animals , CD4 Antigens , CD8 Antigens , Lymphocyte Count , Receptors, Antigen, T-Cell, gamma-delta
7.
Rev Bras Ginecol Obstet ; 43(5): 368-373, 2021 May.
Article in English | MEDLINE | ID: mdl-34077988

ABSTRACT

OBJECTIVE: To evaluate the antitumoral role of γδ TDC cells and αß TDC cells in an experimental model of breast cancer. METHODS: Thirty female Balb/c mice were divided into 2 groups: control group (n = 15) and induced-4T1 group (n = 15), in which the mice received 2 × 105 4T1 mammary tumor cell line. Following the 28-day experimental period, immune cells were collected from the spleen and analyzed by flow cytometry for comparison of αß TDC (TCRαß+ CD11c+MHCII+) and γδ TDC (TCRγδ+CD11c+MHCII+) cells regarding surface markers (CD4+ and C8+) and cytokines (IFN-γ, TNF-α, IL-12 and IL-17). RESULTS: A total of 26.53% of γδ TDC - control group (p < 0.0001) - the proportion of αß TDC was lower in splenic cells than γδ TDC; however, these 2 cell types were reduced in tumor conditions (p < 0.0001), and the proportion of IFN-γ, TNF-α, IL-12 and IL-17 cytokines produced by γδ TDC was higher than those produced by αß TDC, but it decreased under conditions of tumor-related immune system response (p < 0.0001). CONCLUSION: Healthy mice engrafted with malignant cells 4T1 breast tumor presented TDC with γδ TCR repertoire. These cells express cytotoxic molecules of lymphocytes T, producing anti-tumor proinflammatory cytokines.


OBJETIVO: Esclarecer o possível papel antitumoral das células TDC γδ e TDC αß em um modelo experimental de câncer de mama. MéTODOS: Trinta baços de camundongos Balb/c analisados por citometria de fluxo, separados entre grupo controle (n = 15) e o grupo tumoral induzido por 4T1 (n = 15). RESULTADOS: Presença de 26,53% de TDC γδ nos camundongos do grupo controle (p < 0,0001), proporção de TDC αß menor em células esplênicas do que TDC γδ; no entanto, estes dois tipos de células são reduzidos em condições tumorais (p < 0,0001), e a proporção de citocinas IFN-γ, TNF-α, IL-12 e IL-17 produzidas pelas célula TDC γδ foi maior do que as produzidas pelas células TDC αß, mas foram diminuídas sob condições de resposta ao sistema imunológico relacionada ao tumor (p < 0,0001). CONCLUSãO: Camundongos saudáveis induzidos ao tumor de mama 4T1 apresentaram TDC com repertório TCR γδ. Estas células expressam moléculas citotóxicas de linfócitos T, produzindo citocinas proinflamatórias anti-tumor.


Subject(s)
Breast Neoplasms , Receptors, Antigen, T-Cell, alpha-beta , Receptors, Antigen, T-Cell, gamma-delta , Animals , Breast Neoplasms/immunology , Female , Flow Cytometry , Interleukin-17 , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Spleen/immunology , Spleen/metabolism
8.
J Immunol Res ; 2021: 6673722, 2021.
Article in English | MEDLINE | ID: mdl-33954206

ABSTRACT

Invariant Natural Killer T (iNKT) cells are key players in the immunity to several pathogens; however, their involvement in the resistance to Paracoccidioides brasiliensis infection remains unknown. Using splenocytes from CD1d (CD1d-/-) and iNKT-deficient (Jα18-/-) mice, we found that iNKT cells are the innate source of IFN-γ after P. brasiliensis infection and are required to potentiate macrophage oxidative burst and control fungal growth. To determine whether iNKT cells contribute in vivo to host resistance against P. brasiliensis infection, we infected intratracheally wild-type and Jα18-/- C57BL/6 mouse strains with the virulent Pb18 isolate. iNKT cell deficiency impaired the airway acute inflammatory response, resulting in decreased airway neutrophilia and reduced IFN-γ, KC, and nitric oxide (NO) production. The deficient innate immune response of Jα18-/- mice to Pb18 infection resulted in increased fungal burden in the lungs and spleen. Besides, the activation of iNKT cells in vivo by administration of the exogenous iNKT ligand α-galactosylceramide (α-GalCer) improved host resistance to P. brasiliensis infection. Although the mechanisms responsible for this phenomenon remain to be clarified, α-GalCer treatment boosted the local inflammatory response and reduced pulmonary fungal burden. In conclusion, our study is the first evidence that iNKT cells are important for the protective immunity to P. brasiliensis infection and their activation by an exogenous ligand is sufficient to improve the host resistance to this fungal infection.


Subject(s)
Disease Resistance , Natural Killer T-Cells/immunology , Paracoccidioides/immunology , Paracoccidioidomycosis/immunology , Animals , Antigens, CD1d/genetics , Disease Models, Animal , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Paracoccidioidomycosis/microbiology , Receptors, Antigen, T-Cell, alpha-beta/genetics
9.
Rev. bras. ginecol. obstet ; Rev. bras. ginecol. obstet;43(5): 368-373, May 2021. graf
Article in English | LILACS | ID: biblio-1288556

ABSTRACT

Abstract Objective To evaluate the antitumoral role of γδ TDC cells and αβ TDC cells in an experimental model of breast cancer. Methods Thirty female Balb/c mice were divided into 2 groups: control group (n=15) and induced-4T1 group (n=15), in which the mice received 2 x 105 4T1 mammary tumor cell line. Following the 28-day experimental period, immune cells were collected from the spleen and analyzed by flow cytometry for comparison of αβ TDC (TCRαβ+ CD11c+MHCII+) and γδ TDC (TCRγδ+CD11c+MHCII+) cells regarding surface markers (CD4+ and C8+) and cytokines (IFN-γ, TNF-α, IL-12 and IL-17). Results A total of 26.53% of γδ TDC- control group (p<0.0001) - the proportion of αβ TDC was lower in splenic cells than γδ TDC; however, these 2 cell types were reduced in tumor conditions (p<0.0001), and the proportion of IFN-γ, TNF-α, IL-12 and IL-17 cytokines produced by γδ TDC was higher than those produced by αβ TDC, but it decreased under conditions of tumor-related immune system response (p<0.0001). Conclusion Healthy mice engrafted with malignant cells 4T1 breast tumor presented TDC with γδ TCR repertoire. These cells express cytotoxic molecules of lymphocytes T, producing anti-tumor proinflammatory cytokines.


Resumo Objetivo Esclarecer o possível papel antitumoral das células TDC γδ e TDC αβ em um modelo experimental de câncer de mama. Métodos Trinta baços de camundongos Balb/c analisados por citometria de fluxo, separados entre grupo controle (n=15) e o grupo tumoral induzido por 4T1 (n=15). Resultados Presença de 26,53% de TDC γδ nos camundongos do grupo controle (p<0,0001), proporção de TDC αβ menor em células esplênicas do que TDC γδ; no entanto, estes dois tipos de células são reduzidos emcondições tumorais (p<0,0001), e a proporção de citocinas IFN-γ, TNF-α, IL-12 e IL-17 produzidas pelas célula TDC γδ foi maior do que as produzidas pelas células TDC αβ, mas foram diminuídas sob condições de resposta ao sistema imunológico relacionada ao tumor (p<0,0001). Conclusão Camundongos saudáveis induzidos ao tumor de mama 4T1 apresentaram TDC com repertório TCR γδ. Estas células expressam moléculas citotóxicas de linfócitos T, produzindo citocinas proinflamatórias anti-tumor.


Subject(s)
Animals , Female , Mice , Breast Neoplasms/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/immunology , Spleen/metabolism , Interleukin-17 , Flow Cytometry , Mice, Inbred BALB C
10.
Clin Transl Oncol ; 23(8): 1646-1656, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33583004

ABSTRACT

BACKGROUND: Identifying valid biomarkers for patient selection impressively promotes the success of anti-PD-1 therapy. However, the unmet need for biomarkers in gastrointestinal (GI) cancers remains significant. We aimed to explore the predictive value of the circulating T-cell receptor (TCR) repertoire for clinical outcomes in GI cancers who received anti-PD-1 therapy. METHODS: 137 pre- and 79 post-treated peripheral blood samples were included. The TCR repertoire was evaluated by sequencing of complementarity-determining region 3 (CDR3) in the TRB gene. The Shannon index was used to measure the diversity of the TCR repertoire, and Morisita's overlap index was used to determine TCR repertoire similarities between pre- and post-treated samples. RESULTS: Among all enrolled patients, 76 received anti-PD-1 monotherapy and 61 received anti-PD-1 combination therapy. In the anti-PD-1 monotherapy cohort, patients with higher baseline TCR diversity exhibited a significantly higher disease control rate (77.8% vs. 47.2%; hazard ratio [HR] 3.92; 95% confidence interval [CI] 1.14-13.48; P = 0.030) and a longer progression-free survival (PFS) (median: 6.47 months vs. 2.77 months; HR 2.10; 95% CI 1.16-3.79; P = 0.014) and overall survival (OS) (median: NA vs. 8.97 months; HR 3.53; 95% CI 1.49-8.38; P = 0.004) than those with lower diversity. Moreover, patients with a higher TCR repertoire similarity still showed a superior PFS (4.43 months vs. 1.84 months; HR 13.98; 95% CI 4.37-44.68; P < 0.001) and OS (13.40 months vs. 6.12 months; HR 2.93; 95% CI 1.22-7.03; P = 0.016) even in the cohort with lower baseline diversity. However, neither biomarker showed predictive value in the anti-PD-1 combination therapy cohort. Interestingly, the combination of TCR diversity and PD-L1 expression can facilitate patient stratification in a pooled cohort. CONCLUSION: The circulating TCR repertoire can serve as a predictor of clinical outcomes in anti-PD-1 therapy in GI cancers.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Gastrointestinal Neoplasms/therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/blood , Adult , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/blood , Complementarity Determining Regions/genetics , Female , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/mortality , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Progression-Free Survival , Receptors, Antigen, T-Cell, alpha-beta/blood , Time Factors , Treatment Outcome , Young Adult
11.
Front Immunol ; 11: 1279, 2020.
Article in English | MEDLINE | ID: mdl-32695105

ABSTRACT

Chagas disease is an endemic chronic parasitosis in Latin America affecting more than 7 million people. Around 100 million people are currently at risk of acquiring the infection; however, no effective vaccine has been developed yet. Trypanosoma cruzi is the etiological agent of this parasitosis and as an intracellular protozoan it can reside within different tissues, mainly muscle cells, evading host immunity and allowing progression towards the chronic stage of the disease. Considering this intracellular parasitism triggers strong cellular immunity that, besides being necessary to limit infection, is not sufficient to eradicate the parasite from tissues, a differential immune response is required and new strategies for vaccines against Chagas disease need to be explored. In this work, we designed, cloned and expressed a chimeric molecule, named NCz-SEGN24A, comprising a parasite antigen, the N-terminal domain of the major cysteine protease of T. cruzi, cruzipain (Nt-Cz), and a non-toxic form of the staphylococcal superantigen (SAg) G, SEG, with the residue Asn24 mutated to Ala (N24A). The mutant SAg SEGN24A, retains its ability to trigger classical activation of macrophages without inducing T cell apoptosis. To evaluate, as a proof of concept, the immunogenicity and efficacy of the chimeric immunogen vs. its individual antigens, C3H mice were immunized intramuscularly with NCz-SEGN24A co-adjuvanted with CpG-ODN, or the recombinant proteins Nt-Cz plus SEGN24A with the same adjuvant. Vaccinated mice significantly produced Nt-Cz-specific IgG titers after immunization and developed higher IgG2a than IgG1 titers. Specific cell-mediated immunity was assessed by in-vivo DTH and significant responses were obtained. To assess protection, mice were challenged with trypomastigotes of T. cruzi. Both schemes reduced the parasite load throughout the acute phase, but only mice immunized with NCz-SEGN24A showed significant differences against control; moreover, these mice maintained 100% survival. These results encourage testing mutated superantigens fused to specific antigens as immune modulators against pathogens.


Subject(s)
Antigens, Bacterial/immunology , Chagas Disease/prevention & control , Cross Protection/immunology , Cysteine Endopeptidases/immunology , Protozoan Proteins/immunology , Superantigens/immunology , Trypanosoma cruzi/immunology , Animals , Antibodies, Neutralizing , Antibodies, Protozoan/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Protozoan/immunology , Chagas Disease/immunology , Chagas Disease/parasitology , Cysteine Endopeptidases/genetics , Disease Models, Animal , Immunity, Cellular , Immunity, Humoral , Immunization , Mice , Parasite Load , Protein Conformation , Protein Domains/immunology , Protozoan Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Superantigens/chemistry , Superantigens/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Front Immunol ; 11: 953, 2020.
Article in English | MEDLINE | ID: mdl-32508833

ABSTRACT

Background: Visceral leishmaniasis/HIV-co-infected patients (VL/HIV) accounts for around 8% of VL reported cases in Brazil. Relapses of Leishmania infection after anti-leishmanial treatment constitute a great challenge in the clinical practice because of the disease severity and drug resistance. We have shown that non-relapsing-VL/HIV (NR-) evolved with increase of CD4+ T-cell counts and reduction of activated CD4+ and CD8+ T cells after anti-leishmanial treatment. This immune profile was not observed in relapsing-VL/HIV patients (R-), indicating a more severe immunological compromising degree. Elevated activation status may be related to a deficient immune reconstitution and could help to explain the frequent relapses in VL/HIV co-infection. Our aim was to evaluate if this gain of T cells was related to changes in the peripheral TCRVß repertoire and inflammatory status, as well as the possible thymus involvement in the replenishment of these newly formed T lymphocytes. Methods: VL/HIV patients, grouped into non-relapsing (NR- = 6) and relapsing (R- = 12) were evaluated from the active phase up to 12 months post-treatment (mpt). HIV-infected patients (non-VL) and healthy subjects (HS) were included. The TCRVß repertoire was evaluated ex vivo by flow cytometry, whereas the plasmatic cytokine levels were assessed by Luminex assay. To evaluate the thymic output, DNA was extracted from PBMCs for TCR rearrangement excision circles (TREC) quantification by qPCR. Results: VL/HIV cases presented an altered mobilization profile (expansions or retractions) of the TCRVß families when compared to HS independent of the follow-up phase (p < 0.05). TCRVß repertoire on CD4+ T-cells was more homogeneous in the NR-VL/HIV cases, but heterogeneous on CD8+ T-cells, since different Vß-families were mobilized. NR-VL/HIV had the inflammatory pattern reduced after 6 mpt. Importantly, VL/HIV patients showed number of TREC copies lower than controls during all follow-up. An increase of recent thymic emigrants was observed in NR-VL/HIV individuals at 10 mpt compared to R- patients (p < 0.01), who maintained lower TREC contents than the HIV controls. Conclusions: VL/HIV patients that maintain the thymic function, thus generating new T-cells, seem able to replenish the T lymphocyte compartment with effector cells, then enabling parasite control.


Subject(s)
Coinfection , HIV Infections/immunology , Leishmaniasis, Visceral/immunology , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , CD4-CD8 Ratio , Case-Control Studies , Cell Proliferation , Cytokines/blood , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/virology , Humans , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Phenotype , Prospective Studies , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recurrence , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/virology , Thymus Gland/parasitology , Thymus Gland/virology , Time Factors , Treatment Outcome
13.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32526772

ABSTRACT

The monoallelic expression of antigen receptor (AgR) genes, called allelic exclusion, is fundamental for highly specific immune responses to pathogens. This cardinal feature of adaptive immunity is achieved by the assembly of a functional AgR gene on one allele, with subsequent feedback inhibition of V(D)J recombination on the other allele. A range of epigenetic mechanisms have been implicated in sequential recombination of AgR alleles; however, we now demonstrate that a genetic mechanism controls this process for Tcrb. Replacement of V(D)J recombinase targets at two different mouse Vß gene segments with a higher quality target elevates Vß rearrangement frequency before feedback inhibition, dramatically increasing the frequency of T cells with TCRß chains derived from both Tcrb alleles. Thus, TCRß allelic exclusion is enforced genetically by the low quality of Vß recombinase targets that stochastically restrict the production of two functional rearrangements before feedback inhibition silences one allele.


Subject(s)
Alleles , Protein Sorting Signals , Receptors, Antigen, T-Cell, alpha-beta/genetics , V(D)J Recombination/genetics , Animals , Base Sequence , Feedback, Physiological , Gene Expression Regulation , Hybridomas , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , T-Lymphocytes/cytology , Thymocytes/cytology
14.
Viruses ; 12(2)2020 02 20.
Article in English | MEDLINE | ID: mdl-32093256

ABSTRACT

T cells play an essential role in the immune response against the human respiratory syncytial virus (hRSV). It has been described that both CD4+ and CD8+ T cells can contribute to the clearance of the virus during an infection. However, for some individuals, such an immune response can lead to an exacerbated and detrimental inflammatory response with high recruitment of neutrophils to the lungs. The receptor of most T cells is a heterodimer consisting of α and ß chains (αßTCR) that upon antigen engagement induces the activation of these cells. The αßTCR molecule displays a broad sequence diversity that defines the T cell repertoire of an individual. In our laboratory, a recombinant Bacille Calmette-Guérin (BCG) vaccine expressing the nucleoprotein (N) of hRSV (rBCG-N-hRSV) was developed. Such a vaccine induces T cells with a Th1 polarized phenotype that promote the clearance of hRSV infection without causing inflammatory lung damage. Importantly, as part of this work, the T cell receptor (TCR) repertoire of T cells expanded after hRSV infection in naïve and rBCG-N-hRSV-immunized mice was characterized. A more diverse TCR repertoire was observed in the lungs from rBCG-N-hRSV-immunized as compared to unimmunized hRSV-infected mice, suggesting that vaccination with the recombinant rBCG-N-hRSV vaccine triggers the expansion of T cell populations that recognize more viral epitopes. Furthermore, differential expansion of certain TCRVß chains was found for hRSV infection (TCRVß+8.3 and TCRVß+5.1,5.2) as compared to rBCG-N-hRSV vaccination (TCRVß+11 and TCRVß+12). Our findings contribute to better understanding the T cell response during hRSV infection, as well as the functioning of a vaccine that induces a protective T cell immunity against this virus.


Subject(s)
BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Nucleocapsid Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Respiratory Syncytial Virus, Human/immunology , Animals , BCG Vaccine/genetics , Immunity, Cellular , Lung/immunology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins/administration & dosage , Receptors, Antigen, T-Cell/classification , Receptors, Antigen, T-Cell, alpha-beta/genetics , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Specific Pathogen-Free Organisms , Vaccines, Synthetic/immunology
15.
Front Immunol ; 10: 2213, 2019.
Article in English | MEDLINE | ID: mdl-31620131

ABSTRACT

The CSF-470 cellular vaccine plus BCG and rhGM-CSF increased distant metastases-free survival in cutaneous melanoma patients stages IIB-IIC-III relative to medium dose IFN-α2b (CASVAC-0401 study). Patient-045 developed a mature vaccination site (VAC-SITE) and a regional cutaneous metastasis (C-MTS), which were excised during the protocol, remaining disease-free 36 months from vaccination start. CDR3-TCRß repertoire sequencing in PBMC and tissue samples, along with skin-DTH score and IFN-γ ELISPOT assay, were performed to analyze the T-cell immune response dynamics throughout the immunization protocol. Histopathological analysis of the VAC-SITE revealed a highly-inflamed granulomatous structure encircled by CD11c+ nested-clusters, brisk CD8+ and scarce FOXP3+, lymphocytes with numerous Langhans multinucleated-giant-cells and macrophages. A large tumor-regression area fulfilled the C-MTS with brisk lymphocyte infiltration, mainly composed of CD8+PD1+ T-cells, CD20+ B-cells, and scarce FOXP3+ cells. Increasing DTH score and IFN-γ ELISPOT assay signal against the CSF-470 vaccine-lysate was evidenced throughout immunization. TCRß repertoire analysis revealed for the first time the presence of common clonotypes between a VAC-SITE and a C-MTS; most of them persisted in blood by the end of the immunization protocol. In vitro boost with vaccine-lysate revealed the expansion of persistent clones that infiltrated the VAC-SITE and/or the C-MTS; other persistent clones expanded in the patient's blood as well. We propose that expansion of such persistent clonotypes might derive from two different although complementary mechanisms: the proliferation of specific clones as well as the expansion of redundant clones, which increased the number of nucleotide rearrangements per clonotype, suggesting a functional antigenic selection. In this patient, immunization with the CSF-470 vaccine plus BCG and rhGM-CSF induced a T-cell repertoire at the VAC-SITE that was able to infiltrate an emerging C-MTS, which resulted in the expansion of a T-cell repertoire that persisted in blood by the end of the 2-year treatment.


Subject(s)
Cancer Vaccines/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/drug therapy , Melanoma/immunology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , BCG Vaccine/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Male , Middle Aged , Receptors, Antigen, T-Cell, alpha-beta/immunology , Melanoma, Cutaneous Malignant
16.
Front Immunol ; 10: 2080, 2019.
Article in English | MEDLINE | ID: mdl-31555288

ABSTRACT

T-cell receptors (TCR) mediate immune responses recognizing peptides in complex with major histocompatibility complexes (pMHC) displayed on the surface of cells. Resolving the challenge of predicting the cognate pMHC target of a TCR would benefit many applications in the field of immunology, including vaccine design/discovery and the development of immunotherapies. Here, we developed a model for prediction of TCR targets based on similarity to a database of TCRs with known targets. Benchmarking the model on a large set of TCRs with known target, we demonstrated how the predictive performance is increased (i) by focusing on CDRs rather than the full length TCR protein sequences, (ii) by incorporating information from paired α and ß chains, and (iii) integrating information for all 6 CDR loops rather than just CDR3. Finally, we show how integration of the structure of CDR loops, as obtained through homology modeling, boosts the predictive power of the model, in particular in situations where no high-similarity TCRs are available for the query. These findings demonstrate that TCRs that bind to the same target also share, to a very high degree, sequence, and structural features. This observation has profound impact for future development of prediction models for TCR-pMHC interactions and for the use of such models for the rational design of T cell based therapies.


Subject(s)
Complementarity Determining Regions/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Animals , Complementarity Determining Regions/chemistry , Humans , Mice , Peptides , Protein Binding , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Reproducibility of Results , Structure-Activity Relationship
17.
Braz J Infect Dis ; 23(3): 151-159, 2019.
Article in English | MEDLINE | ID: mdl-31271732

ABSTRACT

BACKGROUND: HIV infection harms adaptive cellular immunity mechanisms. Long-term virological control by combined antiretroviral therapy (cART) reduces the risk of mycobacterial infections. Thus, we aimed to study cellular responses to mycobacterial antigens in 20 HIV-infected adolescents with at least one year of virological control (HIV-RNA <40 copies/mL) and 20 healthy adolescents. METHODS: We evaluated CD8 and γδ T-cell degranulation by measurement of CD107a membrane expression after stimulation with lysates from BCG (10 µg/mL) and H37RA Mycobacterium tuberculosis (Mtb, 10 µg/mL). Immune activation and antigen-presenting ability were also assessed by determination of HLA-DR, CD80, and CD86 markers. RESULTS: TCR γδ T-cell CD107a expression was similar between groups in response to mycobacterial antigens, and lower in the HIV-infected group in response to mitogen. Higher baseline HLA-DR expression and lower mycobacterial-stimulated expression was found within the HIV-infected group. CONCLUSIONS: Similar degranulation in stimulated CD8+ and TCR γδ T-cells from HIV-infected adolescents, when compared to healthy controls suggests long-term immunological preservation with immune reconstitution under successful cART. However, differences in HLA-DR expression may represent ongoing inflammation and lower specific responses in HIV-infected youth. These features may be relevant in the context of the precocity and severity of vertically acquired HIV infection.


Subject(s)
AIDS-Related Opportunistic Infections/immunology , Anti-HIV Agents/therapeutic use , Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Tuberculosis/immunology , Antigen Presentation/immunology , Antigens, Bacterial/drug effects , Biomarkers/blood , Cross-Sectional Studies , Female , Humans , Immunophenotyping , Infectious Disease Transmission, Vertical , Male , Prospective Studies , Young Adult
18.
Braz. j. infect. dis ; Braz. j. infect. dis;23(3): 151-159, May-June 2019. tab
Article in English | LILACS | ID: biblio-1019551

ABSTRACT

ABSTRACT Background: HIV infection harms adaptive cellular immunity mechanisms. Long-term virological control by combined antiretroviral therapy (cART) reduces the risk of mycobacterial infections. Thus, we aimed to study cellular responses to mycobacterial antigens in 20 HIV-infected adolescents with at least one year of virological control (HIV-RNA <40 copies/mL) and 20 healthy adolescents. Methods: We evaluated CD8 and γδ T-cell degranulation by measurement of CD107a membrane expression after stimulation with lysates from BCG (10 µg/mL) and H37RA Mycobacterium tuberculosis (Mtb, 10 µg/mL). Immune activation and antigen-presenting ability were also assessed by determination of HLA-DR, CD80, and CD86 markers. Results: TCR γδ T-cell CD107a expression was similar between groups in response to mycobacterial antigens, and lower in the HIV-infected group in response to mitogen. Higher baseline HLA-DR expression and lower mycobacterial-stimulated expression was found within the HIV-infected group. Conclusions: Similar degranulation in stimulated CD8+ and TCR γδ T-cells from HIV-infected adolescents, when compared to healthy controls suggests long-term immunological preservation with immune reconstitution under successful cART. However, differences in HLA-DR expression may represent ongoing inflammation and lower specific responses in HIV-infected youth. These features may be relevant in the context of the precocity and severity of vertically acquired HIV infection.


Subject(s)
Humans , Male , Female , Young Adult , Receptors, Antigen, T-Cell, alpha-beta/immunology , AIDS-Related Opportunistic Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Anti-HIV Agents/therapeutic use , Mycobacterium tuberculosis/immunology , Antigens, Bacterial/immunology , Tuberculosis/immunology , Biomarkers/blood , Cross-Sectional Studies , Prospective Studies , Immunophenotyping , Antigen Presentation/immunology , Infectious Disease Transmission, Vertical , Antigens, Bacterial/drug effects
19.
Front Immunol ; 9: 2291, 2018.
Article in English | MEDLINE | ID: mdl-30337929

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic virus associated with the development of aggressive and poor-prognosis B-cell lymphomas in patients infected with human immunodeficiency virus (HIV+ patients). The most important risk factors for these malignancies include immune dysfunction, chronic immune activation, and loss of T-cell receptor (TCR) repertoire. The combination of all these factors can favor the reactivation of EBV, malignant cell transformation, and clinical progression toward B-cell lymphomas. The overarching aim of this study was to evaluate the frequency, phenotype, functionality, and distribution of TCR clonotypes for EBV-specific T-cell subpopulations in HIV+ patients at different clinical stages and for HIV+ patients with B-cell lymphoma, as well as to establish their association with clinical variables of prognostic value. Factors were studied in 56 HIV+ patients at different clinical stages and in six HIV+ subjects with diagnosed B-cell lymphoma. We found a significant decrease in all subpopulations of EBV-specific CD4+ T cells from HIV+ patients at stage 3 and with B-cell lymphoma. EBV-specific effector CD8+ T cells, particularly effector memory cells, were also reduced in HIV+ patients with B-cell lymphoma. Interestingly, these cells were unable to produce IFN-γ and lacked multifunctionality in HIV+ patients. The TCR-Vß repertoire, which is key for protection against EBV in healthy individuals, was less diverse in HIV+ patients due to a lower frequency of TCR-Vß2+, Vß4+, Vß7.1+, Vß9+, Vß13.6+, Vß14+, Vß17+, Vß22+ CD4+, Vß14+, and Vß17+ CD8+ T cells. HIV+ patients with positive plasma EBV loads (EBV+HIV+) had a noteworthy decrease in the levels of both TNF-α+ and multifunctional TNF-α+/IL-2+ and TNF-α+/IFN-γ+ CD8+ T cells. Altogether, our findings demonstrate that HIV+ patients have significant alterations in the immune response to EBV (poor-quality immunity) that can favor viral reactivation, escalating the risk for developing EBV-associated B-cell lymphomas.


Subject(s)
Coinfection , Epstein-Barr Virus Infections/immunology , HIV Infections/immunology , Herpesvirus 4, Human/immunology , Host-Pathogen Interactions/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cytokines/metabolism , Disease Progression , Epstein-Barr Virus Infections/mortality , Epstein-Barr Virus Infections/virology , Female , HIV Infections/virology , Humans , Immunomodulation , Male , Middle Aged , Prognosis , Young Adult
20.
Front Immunol ; 9: 955, 2018.
Article in English | MEDLINE | ID: mdl-29774030

ABSTRACT

The allogeneic therapeutic vaccine CSF-470 has demonstrated a significant benefit over medium-dose IFNα2b in the distant metastasis-free survival for stages IIB-IIC-III cutaneous melanoma patients in a randomized phase II/III clinical trial (CASVAC-0401, NCT01729663). At the end of the 2-year CSF-470 immunization protocol, patient #006 developed several lung and one subcutaneous melanoma metastases; this later was excised. In this report, we analyzed the changes throughout vaccination of immune populations in blood and in the tumor tissue, with special focus on the T-cell repertoire. Immunohistochemistry revealed a marked increase in CD8+, CD4+, and CD20+ lymphocytes infiltrating the metastasis relative to the primary tumor. Lymphocytes were firmly attached to dying-tumor cells containing Granzyme-B granules. Whole-exon sequencing assessment indicated a moderate-to-high tumor mutational burden, with BRAFV600E as the main oncogenic driver. Mutational signature presented large numbers of mutations at dipyrimidines, typical of melanoma. Relevant tumor and immune-related genes from the subcutaneous metastasis were addressed by RNA-Seq analysis, revealing expression of typical melanoma antigens and proliferative tumor-related genes. Stimulatory and inhibitory immune transcripts were detected as well as evidence of active T-cell effector function. Peripheral blood monitoring revealed an increase in CD4+ and CD8+ cells by the end of the immunization protocol. By CDR3-T-cell receptor ß (TCRß) sequencing, generation of new clones and an increase in oligoclonality was observed in the peripheral T-cells immune repertoire throughout immunization. A shift, with the expansion of selected preexisting and newly arising clones with reduction of others, was detected in blood. In tumor-infiltrating lymphocytes, prevalent clones (50%) were both new and preexisting that were expanded in blood following CSF-470 immunization. These clones persisted in time, since 2 years after completing the immunization, 51% of the clones present in the metastasis were still detected in blood. This is the first report of the modulation of the TCRß repertoire from a melanoma patient immunized with the CSF-470 vaccine. After immunization, the changes observed in peripheral immune populations as well as in the tumor compartment suggest that the vaccine can induce an antitumor adaptive immune repertoire that can reach tumor lesions and persists in blood for at least 2 years.


Subject(s)
Cancer Vaccines/immunology , Melanoma/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Skin Neoplasms/genetics , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/adverse effects , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Female , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Melanoma/therapy , Middle Aged , Mutation , Neoplasm Metastasis , Randomized Controlled Trials as Topic , Sequence Analysis, RNA , Skin Neoplasms/immunology , Vaccination , Melanoma, Cutaneous Malignant
SELECTION OF CITATIONS
SEARCH DETAIL