4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Impaired Thymic Output Can Be Related to the Low Immune Reconstitution and T Cell Repertoire Disturbances in Relapsing Visceral Leishmaniasis Associated HIV/AIDS Patients

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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 TCRVβ 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 TCRVβ 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 TCRVβ families when compared to HS independent of the follow-up phase ( p < 0.05). TCRVβ repertoire on CD4 + T-cells was more homogeneous in the NR-VL/HIV cases, but heterogeneous on CD8 + T-cells, since different Vβ-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.

          Related collections

          Most cited references43

          • Record: found
          • Abstract: found
          • Article: not found

          Changes in thymic function with age and during the treatment of HIV infection.

          The thymus represents the major site of the production and generation of T cells expressing alphabeta-type T-cell antigen receptors. Age-related involution may affect the ability of the thymus to reconstitute T cells expressing CD4 cell-surface antigens that are lost during HIV infection; this effect has been seen after chemotherapy and bone-marrow transplantation. Adult HIV-infected patients treated with highly active antiretroviral therapy (HAART) show a progressive increase in their number of naive CD4-positive T cells. These cells could arise through expansion of existing naive T cells in the periphery or through thymic production of new naive T cells. Here we quantify thymic output by measuring the excisional DNA products of TCR-gene rearrangement. We find that, although thymic function declines with age, substantial output is maintained into late adulthood. HIV infection leads to a decrease in thymic function that can be measured in the peripheral blood and lymphoid tissues. In adults treated with HAART, there is a rapid and sustained increase in thymic output in most subjects. These results indicate that the adult thymus can contribute to immune reconstitution following HAART.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Immune Activation, Inflammation, and Non-AIDS Co-Morbidities in HIV-Infected Patients under Long-Term ART

            Despite effective antiretroviral therapy (ART), people living with HIV (PLWH) still present persistent chronic immune activation and inflammation. This condition is the result of several factors including thymic dysfunction, persistent antigen stimulation due to low residual viremia, microbial translocation and dysbiosis, caused by the disruption of the gut mucosa, co-infections, and cumulative ART toxicity. All of these factors can create a vicious cycle that does not allow the full control of immune activation and inflammation, leading to an increased risk of developing non-AIDS co-morbidities such as metabolic syndrome and cardiovascular diseases. This review aims to provide an overview of the most recent data about HIV-associated inflammation and chronic immune exhaustion in PLWH under effective ART. Furthermore, we discuss new therapy approaches that are currently being tested to reduce the risk of developing inflammation, ART toxicity, and non-AIDS co-morbidities.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Accelerated immune senescence and HIV-1 infection.

              A recent consensus has emerged regarding the association between chronic immune activation and poor outcome in HIV-1 infection. However, its basis remains unclear. Accumulating evidence suggests that the cells of the immune system may have a limited replicative lifespan in vivo. In this context, persistent activation during chronic HIV infection may lead to an exhaustion of immune resources. This may occur at two levels: Clonal and Global. Some HIV-1-specific CD8+ T-cells start expressing the senescence marker CD57 soon after primary infection. Persistently activated HIV-1-specific T-cell clones may eventually reach stages of replicative senescence and disappear, resulting in the specific loss of CD8+ T-cell populations important to control viral replication. In addition, HIV-1 infected individuals are characterized by the accumulation of highly differentiated CD8+ and CD4+ T-cells overtime. Together with the decline of T-cell renewal capacities, this may reflect a general ageing of the lymphocyte population. Similar observations have been done in HIV non-infected elderly individuals, which suggests that premature immunosenescence occurs in HIV-1 infection, as a result of persistent immune activation.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                20 May 2020
                2020
                : 11
                : 953
                Affiliations
                [1] 1Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ) , Rio de Janeiro, Brazil
                [2] 2Núcleo de Ciências Biomédicas Aplicadas, Instituto Federal de Educação, Ciência e Tecnologia Do Rio de Janeiro (IFRJ) , Rio de Janeiro, Brazil
                [3] 3Centro de Referência em Leishmanioses, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ) , Belo Horizonte, Brazil
                [4] 4Laboratório de AIDS e Imunologia Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ) , Rio de Janeiro, Brazil
                [5] 5Laboratório de Alta Complexidade, Instituto Nacional de Saúde da Mulher, da Criança e Do Adolescente Fernandes Figueira (IFF), Fundação Oswaldo Cruz (FIOCRUZ) , Rio de Janeiro, Brazil
                [6] 6Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ) , Rio de Janeiro, Brazil
                [7] 7National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ) , Rio de Janeiro, Brazil
                [8] 8Rede de Pesquisas em Saúde Do Estado Do Rio de Janeiro/FAPERJ , Rio de Janeiro, Brazil
                [9] 9Disciplina de Parasitologia/DMIP, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro (UERJ) , Rio de Janeiro, Brazil
                Author notes

                Edited by: Ann Chidgey, Monash University, Australia

                Reviewed by: Naomi Taylor, National Institutes of Health (NIH), United States; Raffaele De Palma, University of Genoa, Italy

                *Correspondence: Alda Maria Da-Cruz alda@ 123456ioc.fiocruz.br
                Joanna Reis Santos-Oliveira joanna.oliveira@ 123456ifrj.edu.br

                This article was submitted to T Cell Biology, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2020.00953
                7251171
                297ab1e6-f8d2-4a55-aae6-6803c5ec82cf
                Copyright © 2020 Silva-Freitas, Corrêa-Castro, Cota, Giacoia-Gripp, Rabello, Teixeira Dutra, Vasconcelos, Savino, Da-Cruz and Santos-Oliveira.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 19 February 2020
                : 23 April 2020
                Page count
                Figures: 5, Tables: 3, Equations: 0, References: 49, Pages: 14, Words: 10259
                Funding
                Funded by: Funda��o Oswaldo Cruz 10.13039/501100006507
                Award ID: PAEFII-IOC-23-FIO-18-2-53
                Funded by: Conselho Nacional de Desenvolvimento Cient�fico e Tecnol�gico 10.13039/501100003593
                Award ID: Universal � 433637/2018-8
                Funded by: Funda��o Carlos Chagas Filho de Amparo � Pesquisa do Estado do Rio de Janeiro 10.13039/501100004586
                Award ID: E-26/202.944/2016
                Categories
                Immunology
                Original Research

                Immunology
                visceral leishmaniasis/hiv-1 co-infection,thymic output,tcrvβ repertoire,relapses,immune response

                Comments

                Comment on this article