Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Eur J Immunol ; 45(8): 2365-76, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25912253

ABSTRACT

Activation induced deaminase (AID) initiates somatic hypermutation and class switch recombination of the Ig genes in antigen-activated B cells, underpinning antibody affinity maturation and isotype switching. AID can also be pathogenic by contributing to autoimmune diseases and oncogenic mutations. Moreover, AID can exert noncanonical functions when aberrantly expressed in epithelial cells. The lack of specific inhibitors prevents therapeutic applications to modulate AID functions. Here, we have exploited our previous finding that the HSP90 molecular chaperoning pathway stabilizes AID in B cells, to test whether HSP90 inhibitors could target AID in vivo. We demonstrate that chronic administration of HSP90 inhibitors decreases AID protein levels and isotype switching in immunized mice. HSP90 inhibitors also reduce disease severity in a mouse model of acute B-cell lymphoblastic leukemia in which AID accelerates disease progression. We further show that human AID protein levels are sensitive to HSP90 inhibition in normal and leukemic B cells, and that HSP90 inhibition prevents AID-dependent epithelial to mesenchymal transition in a human breast cancer cell line in vitro. Thus, we provide proof-of-concept that HSP90 inhibitors indirectly target AID in vivo and that endogenous human AID is widely sensitive to them, which could have therapeutic applications.


Subject(s)
B-Lymphocytes/immunology , Breast Neoplasms/immunology , Cytidine Deaminase/immunology , HSP90 Heat-Shock Proteins/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Animals , B-Lymphocytes/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/immunology , Female , Humans , Mice , Mice, Knockout , Neoplasms, Experimental/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
2.
J Infect Dis ; 208(6): 952-68, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23966657

ABSTRACT

BACKGROUND: Changes in the phenotype and function of Mycobacterium tuberculosis (M. tuberculosis)-specific CD4+ and CD8+ T-cell subsets in response to stage of infection may allow discrimination between active tuberculosis and latent tuberculosis infection. METHODS: A prospective comparison of M. tuberculosis-specific cellular immunity in subjects with active tuberculosis and latent tuberculosis infection, with and without human immunodeficiency virus (HIV) coinfection. Polychromatic flow cytometry was used to measure CD4+ and CD8+ T-cell subset phenotype and secretion of interferon γ (IFN-γ), interleukin 2 (IL-2), and tumor necrosis factor α (TNF-α). RESULTS: Frequencies of CD4+ and CD8+ cells secreting IFN-γ-only, TNF-α-only and dual IFN-γ/TNF-α were greater in active tuberculosis vs latent tuberculosis infection. All M. tuberculosis-specific CD4+ subsets, with the exception of IL-2-only cells, switched from central to effector memory phenotype in active tuberculosis vs latent tuberculosis infection, accompanied by a reduction in IL-7 receptor α (CD127) expression. The frequency of PPDspecific CD4+ TNF-α-only-secreting T cells with an effector phenotype accurately distinguished active tuberculosis from latent tuberculosis infection with an area under the curve of 0.99, substantially more discriminatory than measurement of function alone. CONCLUSIONS: Combined measurement of T-cell phenotype and function defines a highly discriminatory biomarker of tuberculosis disease activity. Unlocking the diagnostic and monitoring potential of this combined approach now requires validation in large-scale prospective studies.


Subject(s)
Immunophenotyping , Latent Tuberculosis/diagnosis , T-Lymphocyte Subsets/immunology , Adult , Biomarkers/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , HIV , HIV Infections/immunology , HIV Infections/microbiology , Humans , Interferon-gamma/blood , Interleukin-2/blood , Latent Tuberculosis/immunology , Male , Middle Aged , Mycobacterium tuberculosis , Phenotype , Prospective Studies , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Tumor Necrosis Factor-alpha/blood
3.
Front Immunol ; 14: 1166059, 2023.
Article in English | MEDLINE | ID: mdl-38077383

ABSTRACT

The COVID pandemic exposed the critical role T cells play in initial immunity, the establishment and maintenance of long term protection, and of durable responsiveness against novel viral variants. A growing body of evidence indicates that adding measures of cellular immunity will fill an important knowledge gap in vaccine clinical trials, likely leading to improvements in the effectiveness of the next generation vaccines against current and emerging variants. In depth cellular immune monitoring in Phase II trials, particularly for high risk populations such as the elderly or immune compromised, should result in better understanding of the dynamics and requirements for establishing effective long term protection. Such analyses can result in cellular immunity correlates that can then be deployed in Phase III studies using appropriate, scalable technologies. Measures of cellular immunity are less established than antibodies as correlates of clinical immunity, and some misconceptions persist about cellular immune monitoring usefulness, cost, complexity, feasibility, and scalability. We outline the currently available cellular immunity assays, review their readiness for use in clinical trials, their logistical requirements, and the type of information each assay generates. The objective is to provide a reliable source of information that could be leveraged to develop a rational approach for comprehensive immune monitoring during vaccine development.


Subject(s)
Antibodies, Viral , Vaccines , Aged , Humans , Antibodies, Neutralizing , Immunity, Cellular , Vaccine Development
4.
Front Immunol ; 13: 923106, 2022.
Article in English | MEDLINE | ID: mdl-36211354

ABSTRACT

First-generation anit-SARS-CoV-2 vaccines were highly successful. They rapidly met an unforeseen emergency need, saved millions of lives, and simultaneously eased the burden on healthcare systems worldwide. The first-generation vaccines, however, focused too narrowly on antibody-based immunity as the sole marker of vaccine trial success, resulting in large knowledge gaps about waning vaccine protection, lack of vaccine robustness to viral mutation, and lack of efficacy in immunocompromised populations. Detailed reviews of first-generation vaccines, including their mode of action and geographical distribution, have been published elsewhere. Second-generation clinical trials must address these gaps by evaluating a broader range of immune markers, including those representing cell-mediated immunity, to ensure the most protective and long-lasting vaccines are brought to market.


Subject(s)
COVID-19 Vaccines , Clinical Trials as Topic , Humans
5.
J Exp Med ; 198(6): 903-11, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12975456

ABSTRACT

The CD8+ T cell response to Epstein-Barr virus (EBV) is well characterized. Much less is known about the evolution of the CD4+ T cell response. Here we show that EBV stimulates a primary burst of effector CD4+ T cells and this is followed by a period of down-regulation. A small population of EBV-specific effector CD4+ T cells survives during the lifelong persistent phase of infection. The EBV-specific effector CD4+ T cells accumulate within a CD27+ CD28+ differentiation compartment during primary infection and remain enriched within this compartment throughout the persistent phase of infection. Analysis of CD4+ T cell responses to individual epitopes from EBV latent and lytic cycle proteins confirms the observation that the majority of the effector cells express both CD27 and CD28, although CD4+ T cells specific for lytic cycle antigens have a greater tendency to express CD45RA than those specific for the latent antigens. In clear contrast, effector CD4+ T cells specific for cytomegalovirus (CMV) accumulate within the CD27- CD28+ and CD27- CD28- compartments. There are striking parallels in terms of the differentiation of CD8+ T cells specific for EBV and CMV. The results challenge current ideas on the definition of memory subsets.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/metabolism , Immunologic Memory , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/physiology , Cytomegalovirus/immunology , Epitopes , Herpesvirus 4, Human/immunology , Humans , Leukocyte Common Antigens/immunology , Lymphocyte Activation , Phenotype , T-Lymphocyte Subsets , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Viral Proteins/immunology
6.
PLoS One ; 11(1): e0146905, 2016.
Article in English | MEDLINE | ID: mdl-26756579

ABSTRACT

HIV co-infection is an important risk factor for tuberculosis (TB) providing a powerful model in which to dissect out defective, protective and dysfunctional Mycobacterium tuberculosis (MTB)-specific immune responses. To identify the changes induced by HIV co-infection we compared MTB-specific CD4+ responses in subjects with active TB and latent TB infection (LTBI), with and without HIV co-infection. CD4+ T-cell subsets producing interferon-gamma (IFN-γ), interleukin-2 (IL-2) and tumour necrosis factor-alpha (TNF-α) and expressing CD279 (PD-1) were measured using polychromatic flow-cytometry. HIV-TB co-infection was consistently and independently associated with a reduced frequency of CD4+ IFN-γ and IL-2-dual secreting T-cells and the proportion correlated inversely with HIV viral load (VL). The impact of HIV co-infection on this key MTB-specific T-cell subset identifies them as a potential correlate of mycobacterial immune containment. The percentage of MTB-specific IFN-γ-secreting T-cell subsets that expressed PD-1 was increased in active TB with HIV co-infection and correlated with VL. This identifies a novel correlate of dysregulated immunity to MTB, which may in part explain the paucity of inflammatory response in the face of mycobacterial dissemination that characterizes active TB with HIV co-infection.


Subject(s)
CD4-Positive T-Lymphocytes/microbiology , HIV Infections/blood , Mycobacterium tuberculosis , Programmed Cell Death 1 Receptor/metabolism , Tuberculosis/blood , Adult , Antigens, Bacterial/metabolism , CD4-Positive T-Lymphocytes/cytology , Coinfection/microbiology , Coinfection/virology , Female , Gene Expression Regulation , HIV Infections/complications , Humans , Immunophenotyping , Interferon-gamma/metabolism , Interleukin-2/metabolism , Latent Tuberculosis/blood , Latent Tuberculosis/complications , Lymphocyte Subsets/microbiology , Male , Middle Aged , Tuberculosis/complications , Tumor Necrosis Factor-alpha/metabolism , Young Adult
7.
Viral Immunol ; 18(3): 513-22, 2005.
Article in English | MEDLINE | ID: mdl-16212530

ABSTRACT

Antigen-primed cytotoxic T lymphocytes (CTL) may express leukocyte immunoglobulin-like receptors (LILRs) and natural killer receptors (NKRs). Published work suggests that expression of some of these receptors confers survival advantage, leading to the idea that cells expressing such receptors may accumulate as an antigen-specific response evolves. Here we tested this hypothesis by analyzing expression of CD85j (also known as LILRB1 or ILT2), KIRs, CD94, and CD161 by Epstein- Barr virus (EBV)-specific CTL during the primary and persistent phases of EBV infection in humans. During primary infection, few EBV-specific CTL expressed these receptors and this proportion was equally low in early persistent infection. Thus, expression of these molecules does not influence capacity to survive downregulation of the primary response. However, in donors persistently infected with EBV for many years, a significantly higher proportion of EBV-specific CTL expressed CD85j and NKRs, suggesting that cells expressing these receptors can accumulate with time. Using FACS analysis, we confirmed, at a single cell level, that expression of CD85j, defined by staining with the antibody VMP55, was associated with reduced capacity of EBV-specific CD8+ T cells to respond to antigen. Thus, in the later stages of persistent infection, protective immunity to EBV may be reduced due to the preferential accumulation of hyporesponsive EBV-specific CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Receptors, Immunologic/metabolism , Antigens, CD/metabolism , Antigens, Surface/metabolism , Antigens, Viral/administration & dosage , Epstein-Barr Virus Infections/etiology , Herpesvirus 4, Human/immunology , Humans , Immune Tolerance , In Vitro Techniques , Infectious Mononucleosis/etiology , Infectious Mononucleosis/immunology , Interferon-gamma/biosynthesis , Lectins, C-Type/metabolism , Leukocyte Immunoglobulin-like Receptor B1 , Models, Immunological , NK Cell Lectin-Like Receptor Subfamily B , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Receptors, KIR , Time Factors
8.
Immun Inflamm Dis ; 3(3): 141-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26417433

ABSTRACT

HIV-infected individuals with severe immunodeficiency are at risk of opportunistic infection (OI). Tuberculosis (TB) may occur without substantial immune suppression suggesting an early and sustained adverse impact of HIV on Mycobacterium tuberculosis (MTB)-specific cell mediated immunity (CMI). This prospective observational cohort study aimed to observe differences in OI-specific and MTB-specific CMI that might underlie this. Using polychromatic flow cytometry, we compared CD4+ responses to MTB, cytomegalovirus (CMV), Epstein-Barr virus (EBV) and Candida albicans in individuals with and without HIV infection. MTB-specific CD4+ T-cells were more polyfunctional than virus specific (CMV/EBV) CD4+ T-cells which predominantly secreted IFN-gamma (IFN-γ) only. There was a reduced frequency of IFN-γ and IL-2 (IL-2)-dual-MTB-specific cells in HIV-infected individuals, which was not apparent for the other pathogens. MTB-specific cells were less differentiated especially compared with CMV-specific cells. CD127 expression was relatively less frequent on MTB-specific cells in HIV co-infection. MTB-specific CD4+ T-cells PD-1 expression was infrequent in contrast to EBV-specific CD4+ T-cells. The variation in the inherent quality of these CD4+ T-cell responses and impact of HIV co-infection may contribute to the timing of co-infectious diseases in HIV infection.

9.
BMJ Open ; 2(2): e000762, 2012.
Article in English | MEDLINE | ID: mdl-22382123

ABSTRACT

BACKGROUND: HIV is the most important risk factor for progression of latent tuberculosis infection (LTBI) to active tuberculosis (TB). Detection and treatment of LTBI is necessary to reduce the increasing burden of TB in the UK, but a unified LTBI screening approach has not been adopted. OBJECTIVE: To compare the effectiveness of a TB risk-focused approach to LTBI screening in the HIV-positive population against current UK National Institute for Health and Clinical Excellence (NICE) guidance. DESIGN: Prospective cohort study. SETTING: Two urban HIV treatment centres in London, UK. PARTICIPANTS: 114 HIV-infected individuals with defined TB risk factors were enrolled prospectively as part of ongoing studies into HIV and TB co-infection. OUTCOME MEASURES: The yield and case detection rate of LTBI cases within the research study were compared with those generated by the NICE criteria. RESULTS: 17/114 (14.9%, 95% CI 8.3 to 21.5) had evidence of LTBI. Limiting screening to those meeting NICE criteria for the general population (n=43) would have detected just over half of these, 9/43 (20.9%, 95% CI 8.3 to 33.5) and those meeting criteria for HIV co-infection (n=74) would only have captured 8/74(10.8%, 95% CI 3.6 to 18.1) cases. The case detection rates from the study and NICE approaches were not significantly different. LTBI was associated with the presence of multiple TB risk factors (p=0.002). CONCLUSION: Adoption of a TB risk-focused screening algorithm that does not use CD4 count stratification could prevent more cases of TB reactivation, without changing the case detection rate. These findings should be used to inform a large-scale study to create unified guidelines.

10.
Nat Prod Commun ; 6(5): 689-94, 2011 May.
Article in English | MEDLINE | ID: mdl-21615033

ABSTRACT

Cyanobacteria (= blue-green algae) are prolific producers of structurally distinct and biologically active metabolites. In the continuation of our search for new sources of anti-infective natural products, we have assessed the in vitro antiprotozoal (Plasmodium falciparum, Trypanosoma brucei rhodesiense, T. cruzi, Leishmania donovani) and antitubercular (Mycobacterium tuberculosis) potential of samples of two terrestrial cyanobacteria, Nostoc commune (collected when desiccated and wet) and Rivularia biasolettiana. The cytotoxic potential of the extracts was also evaluated against primary L6 cells. Except for T. cruzi and M. tuberculosis, the crude extracts were active against all the organisms tested and showed no toxicity. The crude extracts were then partitioned between n-hexane, chloroform and aqueous methanol and retested against the same panel of pathogens. The chloroform sub-extracts of both N. commune samples showed significant activity against T. b. rhodesiense (IC50 values 2.0 and 3.5 microg/mL) and P. falciparum (IC50s 7.4 and 5.8 microg/mL), with low toxicity. This trend was also true for R. biasolettiana extracts, and its chloroform sub-extract showed notable activity against all parasitic protozoa. There were differences in the biological activity profiles of extracts derived from desiccated and hydrated forms of N. commune. To our knowledge, this is the first study assessing the anti-infective activity of desiccated and hydrated forms of N. commune, as well as R. biasolettiana. Furthermore, the present work reports such biological activity in terrestrial cyanobacteria from Ireland for the first time. These results warrant the further study of Irish terrestrial cyanobacteria as a valuable source of new natural product leads for the treatment of parasitic protozoal infections.


Subject(s)
Antineoplastic Agents/analysis , Antiprotozoal Agents/analysis , Antitubercular Agents/analysis , Nostoc commune/chemistry , Animals , Cell Line , Ireland , Microbial Sensitivity Tests , Rats
11.
J Clin Invest ; 121(6): 2493-503, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21576820

ABSTRACT

Current tuberculosis (TB) vaccine strategies are largely aimed at activating conventional T cell responses to mycobacterial protein antigens. However, the lipid-rich cell wall of Mycobacterium tuberculosis (M. tuberculosis) is essential for pathogenicity and provides targets for unconventional T cell recognition. Group 1 CD1-restricted T cells recognize mycobacterial lipids, but their function in human TB is unclear and their ability to establish memory is unknown. Here, we characterized T cells specific for mycolic acid (MA), the predominant mycobacterial cell wall lipid and key virulence factor, in patients with active TB infection. MA-specific T cells were predominant in TB patients at diagnosis, but were absent in uninfected bacillus Calmette-Guérin-vaccinated (BCG-vaccinated) controls. These T cells were CD1b restricted, detectable in blood and disease sites, produced both IFN-γ and IL-2, and exhibited effector and central memory phenotypes. MA-specific responses contracted markedly with declining pathogen burden and, in patients followed longitudinally, exhibited recall expansion upon antigen reencounter in vitro long after successful treatment, indicative of lipid-specific immunological memory. T cell recognition of MA is therefore a significant component of the acute adaptive and memory immune response in TB, suggesting that mycobacterial lipids may be promising targets for improved TB vaccines.


Subject(s)
Antigens, Bacterial/immunology , Antigens, CD1/immunology , Cell Wall/immunology , Immunologic Memory/immunology , Mycobacterium tuberculosis/immunology , Mycolic Acids/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/immunology , Tuberculosis/immunology , Acute Disease , Adaptive Immunity , Adult , Aged , Antitubercular Agents/therapeutic use , BCG Vaccine/immunology , Cells, Cultured/immunology , Female , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Middle Aged , Mycobacterium tuberculosis/pathogenicity , T-Lymphocyte Subsets/metabolism , Tuberculosis/drug therapy , Tuberculosis/prevention & control , Tuberculosis Vaccines , Virulence , Young Adult
12.
PLoS One ; 5(12): e15619, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21179481

ABSTRACT

BACKGROUND: IFN-γ and IL-2 cytokine-profiles define three functional T-cell subsets which may correlate with pathogen load in chronic intracellular infections. We therefore investigated the feasibility of the immunospot platform to rapidly enumerate T-cell subsets by single-cell IFN-γ/IL-2 cytokine-profiling and establish whether immunospot-based T-cell signatures distinguish different clinical stages of human tuberculosis infection. METHODS: We used fluorophore-labelled anti-IFN-γ and anti-IL-2 antibodies with digital overlay of spatially-mapped colour-filtered images to enumerate dual and single cytokine-secreting M. tuberculosis antigen-specific T-cells in tuberculosis patients and in latent tuberculosis infection (LTBI). We validated results against established measures of cytokine-secreting T-cells. RESULTS: Fluorescence-immunospot correlated closely with single-cytokine enzyme-linked-immunospot for IFN-γ-secreting T-cells and IL-2-secreting T-cells and flow-cytometry-based detection of dual IFN-γ/IL-2-secreting T-cells. The untreated tuberculosis signature was dominated by IFN-γ-only-secreting T-cells which shifted consistently in longitudinally-followed patients during treatment to a signature dominated by dual IFN-γ/IL-2-secreting T-cells in treated patients. The LTBI signature differed from active tuberculosis, with higher proportions of IL-2-only and IFN-γ/IL-2-secreting T-cells and lower proportions of IFN-γ-only-secreting T-cells. CONCLUSIONS: Fluorescence-immunospot is a quantitative, accurate measure of functional T-cell subsets; identification of cytokine-signatures of pathogen burden, distinct clinical stages of M. tuberculosis infection and long-term immune containment suggests application for treatment monitoring and vaccine evaluation.


Subject(s)
Microscopy, Fluorescence/methods , T-Lymphocytes/cytology , Tuberculosis/microbiology , Adolescent , Adult , Aged , Case-Control Studies , Cell Count , Cross-Sectional Studies , Female , Flow Cytometry/methods , Humans , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Middle Aged , Mycobacterium tuberculosis/metabolism , Risk Factors , Tuberculosis/blood
13.
J Infect Dis ; 191(11): 1842-53, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15871117

ABSTRACT

Leukocyte immunoglobulin-like receptor-1 (LIR-1) is an inhibitory receptor that negatively regulates T cell effector functions after interaction with host class I major histocompatibility complex molecules and, additionally, binds to UL18, a human cytomegalovirus (HCMV)-encoded class I homologue. Here, we demonstrate that virus-specific cytotoxic T lymphocytes (CTLs) differentially express LIR-1, with high frequencies of expression on HCMV-specific CD8+ T cells and intermediate and low frequencies of expression on influenza virus-specific and Epstein-Barr virus (EBV)-specific CTLs, respectively. Expression of LIR-1 was dependent on CTL-antigen specificity and was associated with a differentiated effector memory phenotype, as demonstrated by decreased expression of CD28 and increased expression of CD57. During primary HCMV and EBV infections, expression of LIR-1 on virus-specific CTLs was low and increased slowly. These results indicate that expression of LIR-1 increases during differentiation of virus-specific CD8+ effector T cells. Furthermore, they suggest that a potential immunoregulatory function of UL18 may be to preferentially target highly differentiated HCMV-specific effector memory T cells during persistent infection.


Subject(s)
Antigens, CD/metabolism , Cytomegalovirus Infections/immunology , Gene Expression Regulation/immunology , Receptors, Immunologic/metabolism , T-Lymphocytes, Cytotoxic/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epstein-Barr Virus Infections/immunology , HLA Antigens/metabolism , Humans , Influenza, Human/immunology , Leukocyte Immunoglobulin-like Receptor B1 , Recurrence , Species Specificity
14.
Curr Immunol Rev ; 9: 72-85, 2013. ilus
Article in English | URUCAN | ID: bcc-5326

ABSTRACT

The enzyme Activation-induced cytidine deaminase (AID) initiates somatic hypermutation (SHM) and class switch recombination (CSR) of the immunoglobulin (Ig) genes, which are critically important for an effective immune response. In addition, AID seems to contribute to B cell tolerance in mice and humans by, in some still undefined way, eliminating developing autoreactive B cells. As a trade-off for the benefits brought about by its physiological roles, AID can also contribute to cellular transformation and tumor progression through its mutagenic activity. AID deaminates deoxycytidines at the Ig genes thereby generating deoxyuridine, which as part of the normal mechanism of SHM and CSR is processed by DNA repair enzymes into a larger spectrum of point mutations and also DNA double-strand breaks. Multiple mechanisms regulate AID function to minimize deleterious or pathogenic DNA damage during antibody gene diversification. Despite this, off-target AID activity still makes point mutations and initiates chromosomal translocations that affect tumor suppressor and proto-oncogenes associated with B-cell lymphoid neoplasms. Through this collateral damage, AID is etiological for the development of lymphoma in several mouse models and is expressed in many human malignancies of mature B-cell origin where it may contribute to tumor clonal evolution. Mounting evidences indicate a role for AID also in disease progression and worsening of the prognosis of Chronic Lymphocytic Leukemia (CLL) and Chronic Myelogenous Leukemia (CML)(cont) (AU)


Subject(s)
Humans , Leukemia, Lymphocytic, Chronic, B-Cell , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Bibliography, National , Uruguay
SELECTION OF CITATIONS
SEARCH DETAIL