Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Thromb Haemost ; 121(11): 1476-1482, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33759145

ABSTRACT

BACKGROUND: Elucidating mechanisms of brain damage in cerebral venous thrombosis (CVT) would be instrumental to develop targeted therapies and improve prognosis prediction. Matrix metalloproteinase-9 (MMP-9), a gelatinase that degrades major components of the basal lamina, has been associated to blood-brain barrier disruption. We aimed to assess, in patients with CVT, the temporal change in serum concentrations of MMP-9 and its association with key imaging and clinical outcomes. METHODS: Pathophysiology of Venous Infarction-PRediction of InfarctiOn and RecanalIzaTion in CVT (PRIORITy-CVT) was a multicenter prospective cohort study of patients with newly diagnosed CVT. Serial collection of peripheral blood samples performed on day 1, 3, and 8, and standardized magnetic resonance imaging on day 1, 8, and 90. MMP-9 was quantified using enzyme-linked immunosorbent assay in 59 patients and 22 healthy controls. Primary outcomes were parenchymal brain lesion, early evolution of brain lesion, early recanalization, and functional outcome on day 90. RESULTS: CVT patients with parenchymal brain lesion had higher baseline concentrations of MMP-9 compared with controls (adjusted p = 0.001). The area under receiver operating characteristic curve value for MMP-9 for predicting brain lesion was 0.71 (95% confidence interval [CI]: 0.57-0.85, p = 0.009). Patients with venous recanalization showed early decline of circulating MMP-9 and significantly lower levels on day 8 (p = 0.021). Higher MMP-9 on day 8 was associated with persistent venous occlusion (odds ratio: 1.20 [per 20 ng/mL], 95% CI: 1.02-1.43, p = 0.030). CONCLUSION: We report a novel relationship among MMP-9, parenchymal brain damage, and early venous recanalization, suggesting that circulating MMP-9 is a dynamic marker of brain tissue damage in patients with CVT.


Subject(s)
Cerebral Veins , Intracranial Thrombosis/enzymology , Matrix Metalloproteinase 9/blood , Venous Thrombosis/enzymology , Adult , Biomarkers/blood , Case-Control Studies , Cerebral Angiography , Cerebral Veins/diagnostic imaging , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intracranial Thrombosis/blood , Intracranial Thrombosis/diagnostic imaging , Magnetic Resonance Angiography , Male , Middle Aged , Phlebography , Portugal , Predictive Value of Tests , Prognosis , Prospective Studies , Time Factors , Venous Thrombosis/blood , Venous Thrombosis/diagnostic imaging , Young Adult
2.
J Infect Dis ; 221(1): 122-126, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31504642

ABSTRACT

Follicular helper T cells (Tfh), CD4 lymphocytes critical for efficient antibody responses, have been shown to be key human immunodeficiency virus (HIV)-1 reservoirs. Human immunodeficiency virus-2 infection represents a unique naturally occurring model for investigating Tfh role in HIV/acquired immune deficiency syndrome, given its slow rate of CD4 decline, low to undetectable viremia, and high neutralizing antibody titers throughout the disease course. In this study, we investigated, for the first time, Tfh susceptibility to HIV-2 infection by combining in vitro infection of tonsillar Tfh with the ex vivo study of circulating Tfh from HIV-2-infected patients. We reveal that Tfh support productive HIV-2 infection and are preferential viral targets in HIV-2-infected individuals.


Subject(s)
DNA, Viral/metabolism , HIV Infections/immunology , HIV-1/physiology , HIV-2/physiology , T-Lymphocytes, Helper-Inducer/virology , Female , HIV Infections/virology , Humans , Middle Aged , Palatine Tonsil/immunology , Palatine Tonsil/pathology , Primary Cell Culture , RNA, Messenger/metabolism , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Viral Tropism , gag Gene Products, Human Immunodeficiency Virus/genetics
3.
Front Immunol ; 10: 2503, 2019.
Article in English | MEDLINE | ID: mdl-31824477

ABSTRACT

Monozygotic twins provide a unique opportunity to better understand complex genetic diseases and the relative contribution of heritable factors in shaping the immune system throughout life. Common Variable Immunodeficiency Disorders (CVID) are primary antibody defects displaying wide phenotypic and genetic heterogeneity, with monogenic transmission accounting for only a minority of the cases. Here, we report a pair of monozygotic twins concordant for CVID without a family history of primary immunodeficiency. They featured a remarkably similar profile of clinical manifestations and immunological alterations at diagnosis (established at age 37) and along the subsequent 15 years of follow-up. Interestingly, whole-exome sequencing failed to identify a monogenic cause for CVID, but unraveled a combination of heterozygous variants, with a predicted deleterious impact. These variants were found in genes involved in relevant immunological pathways, such as JUN, PTPRC, TLR1, ICAM1, and JAK3. The potential for combinatorial effects translating into the observed disease phenotype is inferred from their roles in immune pathways, namely in T and B cell activation. The combination of these genetic variants is also likely to impose a significant constraint on environmental influences, resulting in a similar immunological phenotype in both twins, despite exposure to different living conditions. Overall, these cases stress the importance of integrating NGS data with clinical and immunological phenotypes at the single-cell level, as provided by multi-dimensional flow-cytometry, in order to understand the complex genetic landscape underlying the vast majority of patients with CVID, as well as those with other immunodeficiencies.


Subject(s)
Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/etiology , Disease Susceptibility , Twins, Monozygotic , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Humans , Immunophenotyping , Male , Multifactorial Inheritance , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Symptom Assessment , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Exome Sequencing
4.
Acta Med Port ; 32(7-8): 514-519, 2019 Aug 01.
Article in Portuguese | MEDLINE | ID: mdl-31445531

ABSTRACT

INTRODUCTION: Children with DiGeorge syndrome/chromosome 22q11.2 deletion syndrome might have a variable degree of immunodeficiency, which may limit the use of live vaccines. The aim of this study was to review the adverse effects of live vaccines and possible relation with immune status in patients with DiGeorge Syndrome/partial 22q11.2 deletion syndrome. MATERIAL AND METHODS: Retrospective study with analysis of the clinical records of children with chromosome 22q11.2 deletion syndrome and DiGeorge syndrome phenotype, followed in a Primary Immunodeficiency center. Data were collected on: demographic characteristics; medical and vaccination history with live vaccines; T-CD4+ lymphocyte counts and lymphocyte proliferative responses to antigens and mitogens; adverse reactions; vaccine failure. RESULTS: Twenty three children with DiGeorge syndrome/22q11.2 deletion syndrome were included, 65.2% male, with average age at diagnosis of 11.3 months. Eighteen children (78%) received bacillus Calmette-Guérin vaccine: all with evidence of thymic activity; three presented moderate T-CD4+ lymphopenia and abnormal lymphocyte proliferative responses; one had abnormal lymphocyte proliferative responses for mitogens, four for purified protein derivative and one for tetanus toxoid. Measles, mumps and rubella vaccine was administered to 15 children, three of them with moderate immunosuppression and abnormal lymphocyte proliferative responses. Live attenuated polio vaccine was administered to 4 children without immunosuppression and the rotavirus vaccine to three children, one with moderate immunosuppression. No significant adverse reactions were reported. DISCUSSION: These data are in line with the findings of other international studies. CONCLUSION: In our sample, live vaccines were well-tolerated, even in children with moderate T-CD4+ lymphopenia and abnormal lymphocyte proliferative responses to antigens/mitogens.


Introdução: A síndrome de DiGeorge/deleção 22q11.2 pode apresentar um grau variável de imunodeficiência, condicionando a utilização de vacinas vivas. Este estudo teve como objetivo documentar os efeitos adversos de vacinas vivas e possível relação com alterações imunitárias em crianças com síndrome de DiGeorge/deleção 22q11.2 parcial. Material e Métodos: Foi realizado um estudo retrospetivo por revisão dos processos clínicos das crianças com deleção do cromossoma 22q11.2 e fenótipo de síndrome de DiGeorge, seguidos num centro de referência de imunodeficiências primárias. Foi realizada colheita de dados, incluindo: características demográficas; história médica; historial de vacinação com vacinas vivas; contagem de linfócitos T-CD4+ e respostas proliferativas linfocitárias a antigénios e mitogénios; reações adversas; falências vacinais. Resultados: Foram incluídas 23 crianças com síndrome de DiGeorge/deleção 22q11.2, 65,2% do sexo masculino e idade média de diagnóstico de 11,3 meses. Destas, 18 crianças (78%) receberam a vacina bacillus Calmette-Guérin: todas com evidência de atividade tímica; três apresentaram linfopénia T-CD4+ moderada e respostas proliferativas linfocitárias anormais; uma com respostas proliferativas linfocitárias anormais para mitogénios, quatro para derivado de proteína purificada e uma para toxóide tetânico. A vacina tríplice contra o sarampo, parotidite e rubéola foi administrada a 15 crianças, três com imunossupressão moderada e respostas proliferativas linfocitárias anormais. A vacina viva atenuada contra poliomielite foi administrada a quatro crianças sem imunossupressão e a vacina contra o rotavírus a três crianças, uma com imunossupressão moderada. Não foram reportadas reações adversas. Discussão: Estes dados estão de acordo com as conclusões de outros estudos internacionais. Conclusão: Na nossa amostra, as vacinas vivas atenuadas foram bem toleradas, incluindo em crianças com linfopénia T-CD4+ moderada e com respostas proliferativas linfocitárias a antigénios/mitogénios anormais.


Subject(s)
DiGeorge Syndrome/immunology , Immune Tolerance , Adjuvants, Immunologic/administration & dosage , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes , Cell Proliferation , Child , Child, Preschool , Female , Humans , Immunocompromised Host , Infant , Lymphopenia/immunology , Male , Measles-Mumps-Rubella Vaccine/administration & dosage , Measles-Mumps-Rubella Vaccine/immunology , Phenotype , Poliovirus Vaccine, Inactivated/administration & dosage , Retrospective Studies , Rotavirus Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
5.
J Allergy Clin Immunol ; 144(3): 809-824, 2019 09.
Article in English | MEDLINE | ID: mdl-30826363

ABSTRACT

BACKGROUND: Predominantly antibody deficiencies (PADs) are the most prevalent primary immunodeficiencies, but their B-cell defects and underlying genetic alterations remain largely unknown. OBJECTIVE: We investigated patients with PADs for the distribution of 41 blood B-cell and plasma cell (PC) subsets, including subsets defined by expression of distinct immunoglobulin heavy chain subclasses. METHODS: Blood samples from 139 patients with PADs, 61 patients with common variable immunodeficiency (CVID), 68 patients with selective IgA deficiency (IgAdef), 10 patients with IgG subclass deficiency with IgA deficiency, and 223 age-matched control subjects were studied by using flow cytometry with EuroFlow immunoglobulin isotype staining. Patients were classified according to their B-cell and PC immune profile, and the obtained patient clusters were correlated with clinical manifestations of PADs. RESULTS: Decreased counts of blood PCs, memory B cells (MBCs), or both expressing distinct IgA and IgG subclasses were identified in all patients with PADs. In patients with IgAdef, B-cell defects were mainly restricted to surface membrane (sm)IgA+ PCs and MBCs, with 2 clear subgroups showing strongly decreased numbers of smIgA+ PCs with mild versus severe smIgA+ MBC defects and higher frequencies of nonrespiratory tract infections, autoimmunity, and affected family members. Patients with IgG subclass deficiency with IgA deficiency and those with CVID showed defects in both smIgA+ and smIgG+ MBCs and PCs. Reduced numbers of switched PCs were systematically found in patients with CVID (absent in 98%), with 6 different defective MBC (and clinical) profiles: (1) profound decrease in MBC numbers; (2) defective CD27+ MBCs with almost normal IgG3+ MBCs; (3) absence of switched MBCs; and (4) presence of both unswitched and switched MBCs without and; (5) with IgG2+ MBCs; and (6) with IgA1+ MBCs. CONCLUSION: Distinct PAD defective B-cell patterns were identified that are associated with unique clinical profiles.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunologic Deficiency Syndromes/immunology , Plasma Cells/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Cell Count , Child , Child, Preschool , Female , Humans , Immunoglobulins/deficiency , Immunoglobulins/immunology , Male , Middle Aged , Young Adult
6.
J Allergy Clin Immunol ; 141(6): 2208-2219.e16, 2018 06.
Article in English | MEDLINE | ID: mdl-29505809

ABSTRACT

BACKGROUND: Humoral immunocompetence develops stepwise throughout life and contributes to individual susceptibility to infection, immunodeficiency, autoimmunity, and neoplasia. Immunoglobulin heavy chain (IgH) isotype serum levels can partly explain such age-related differences, but their relationship with the IgH isotype distribution within memory B-cell (MBC) and plasma cell (PCs) compartments remains to be investigated. OBJECTIVE: We studied the age-related distribution of MBCs and PCs expressing different IgH isotypes in addition to the immature/transitional and naive B-cell compartments. METHODS: B-cell and PC subsets and plasma IgH isotype levels were studied in cord blood (n = 19) and peripheral blood (n = 215) from healthy donors aged 0 to 90 years by using flow cytometry and nephelometry, respectively. RESULTS: IgH-switched MBCs expressing IgG1, IgG2, IgG3, IgA1, and IgA2 were already detected in cord blood and newborns at very low counts, whereas CD27+IgM++IgD+ MBCs only became detectable at 1 to 5 months and remained stable until 2 to 4 years, and IgD MBCs peaked at 2 to 4 years, with both populations decreasing thereafter. MBCs expressing IgH isotypes of the second immunoglobulin heavy chain constant region (IGHC) gene block (IgG1, IgG3, and IgA1) peaked later during childhood (2-4 years), whereas MBCs expressing third IGHC gene block immunoglobulin isotypes (IgG2, IgG4, and IgA2) reached maximum values during adulthood. PCs were already detected in newborns, increasing in number until 6 to 11 months for IgM, IgG1, IgG2, IgG3, IgA1, and IgA2; until 2 to 4 years for IgD; and until 5 to 9 years for IgG4 and decreasing thereafter. For most IgH isotypes (except IgD and IgG4), maximum plasma levels were reached after PC and MBC counts peaked. CONCLUSIONS: PC counts reach maximum values early in life, followed by MBC counts and plasma IgH isotypes. Importantly, IgH isotypes from different IGHC gene blocks show different patterns, probably reflecting consecutive cycles of IgH isotype switch recombination through life.


Subject(s)
Aging/immunology , B-Lymphocytes/immunology , Immunity, Humoral/immunology , Immunologic Memory/immunology , Plasma Cells/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Immunoglobulin Class Switching/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Isotypes/blood , Immunoglobulin Isotypes/immunology , Infant , Infant, Newborn , Male , Middle Aged , Young Adult
7.
Oncotarget ; 8(25): 40514-40532, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28465489

ABSTRACT

We propose a novel single-deoxynucleoside-based assay that is easy to perform and provides accurate values for the absolute length (in units of time) of each of the cell cycle stages (G1, S and G2/M). This flow-cytometric assay takes advantage of the excellent stoichiometric properties of azide-fluorochrome detection of DNA substituted with 5-ethynyl-2'-deoxyuridine (EdU). We show that by pulsing cells with EdU for incremental periods of time maximal EdU-coupled fluorescence is reached when pulsing times match the length of S phase. These pulsing times, allowing labelling for a full S phase of a fraction of cells in asynchronous populations, provide accurate values for the absolute length of S phase. We characterized additional, lower intensity signals that allowed quantification of the absolute durations of G1 and G2 phases.Importantly, using this novel assay data on the lengths of G1, S and G2/M phases are obtained in parallel. Therefore, these parameters can be estimated within a time frame that is shorter than a full cell cycle. This method, which we designate as EdU-Coupled Fluorescence Intensity (E-CFI) analysis, was successfully applied to cell types with distinctive cell cycle features and shows excellent agreement with established methodologies for analysis of cell cycle kinetics.


Subject(s)
Cell Cycle , DNA/chemistry , Deoxyuridine/analogs & derivatives , Fluorescence , Cell Division , Click Chemistry/methods , DNA/genetics , Deoxyuridine/chemistry , Flow Cytometry/methods , G1 Phase , G2 Phase , HCT116 Cells , Humans , Hydrazines/chemistry , Kinetics , S Phase , Time Factors
8.
EMBO J ; 36(3): 346-360, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27993935

ABSTRACT

Cell activation is a vital step for T-cell memory/effector differentiation as well as for productive HIV infection. To identify novel regulators of this process, we used next-generation sequencing to profile changes in microRNA expression occurring in purified human naive CD4 T cells in response to TCR stimulation and/or HIV infection. Our results demonstrate, for the first time, the transcriptional up-regulation of miR-34c-5p in response to TCR stimulation in naive CD4 T cells. The induction of this miR was further consistently found to be reduced by both HIV-1 and HIV-2 infections. Overexpression of miR-34c-5p led to changes in the expression of several genes involved in TCR signaling and cell activation, confirming its role as a novel regulator of naive CD4 T-cell activation. We additionally show that miR-34c-5p promotes HIV-1 replication, suggesting that its down-regulation during HIV infection may be part of an anti-viral host response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV/physiology , Host-Pathogen Interactions , Lymphocyte Activation , MicroRNAs/analysis , Receptors, Antigen, T-Cell/metabolism , Virus Replication , CD4-Positive T-Lymphocytes/virology , Gene Expression Profiling , HIV/immunology , Humans , Immune Evasion
9.
Oncotarget ; 7(11): 12163-75, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26910841

ABSTRACT

Naïve FoxP3-expressing regulatory T-cells (Tregs) are essential to control immune responses via continuous replenishment of the activated-Treg pool with thymus-committed suppressor cells. The mechanisms underlying naïve-Treg maintenance throughout life in face of the age-associated thymic involution remain unclear. We found that in adults thymectomized early in infancy the naïve-Treg pool is remarkably well preserved, in contrast to conventional naïve CD4 T-cells. Naïve-Tregs featured high levels of cycling and pro-survival markers, even in healthy individuals, and contrasted with other circulating naïve/memory CD4 T-cell subsets in terms of their strong γc-cytokine-dependent signaling, particularly in response to IL-7. Accordingly, ex-vivo stimulation of naïve-Tregs with IL-7 induced robust cytokine-dependent signaling, Bcl-2 expression, and phosphatidylinositol 3-kinase (PI3K)-dependent proliferation, whilst preserving naïve phenotype and suppressive capacity. Altogether, our data strongly implicate IL-7 in the thymus-independent long-term survival of functional naïve-Tregs, and highlight the potential of targeting the IL-7 pathway to modulate Tregs in different clinical settings.


Subject(s)
Interleukin-7/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Cell Survival/immunology , Forkhead Transcription Factors/immunology , Humans , Young Adult
10.
Haematologica ; 99(6): 1062-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24561792

ABSTRACT

Adult B-cell acute lymphoblastic leukemia remains a major therapeutic challenge, requiring a better characterization of the molecular determinants underlying disease progression and resistance to treatment. Here, using a phospho-flow cytometry approach we show that adult diagnostic B-cell acute lymphoblastic leukemia specimens display PI3K/Akt pathway hyperactivation, irrespective of their BCR-ABL status and despite paradoxically high basal expression of PTEN, the major negative regulator of the pathway. Protein kinase CK2 is known to phosphorylate PTEN thereby driving PTEN protein stabilization and concomitant PTEN functional inactivation. In agreement, we found that adult B-cell acute lymphoblastic leukemia samples show significantly higher CK2 kinase activity and lower PTEN lipid phosphatase activity than healthy controls. Moreover, the clinical-grade CK2 inhibitor CX-4945 (Silmitasertib) reversed PTEN levels in leukemia cells to those observed in healthy controls, and promoted leukemia cell death without significantly affecting normal bone marrow cells. Our studies indicate that CK2-mediated PTEN posttranslational inactivation, associated with PI3K/Akt pathway hyperactivation, are a common event in adult B-cell acute lymphoblastic leukemia and suggest that CK2 inhibition may constitute a valid, novel therapeutic tool in this malignancy.


Subject(s)
PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adolescent , Adult , Aged , Case-Control Studies , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cell Line , Chromosome Aberrations , Enzyme Activation , Female , Gene Expression , Humans , Immunohistochemistry , Janus Kinases/metabolism , Male , Middle Aged , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Kinase Inhibitors/pharmacology , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Young Adult
11.
Immunol Cell Biol ; 88(5): 523-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20142839

ABSTRACT

The thymus generates a T-cell lineage dedicated to immune regulation, 'naturally occurring' regulatory T cells, best specified by the forkhead family transcription factor Foxp3. Here, we have conducted a parallel study in humans and mice where we have dissected the earliest stages of Foxp3 induction during thymocyte development. By analyzing a large collection of 21 human thymuses we show that Foxp3 can be consistently detected in CD4 immature single positive thymocytes that precede the CD4(+)CD8(+) (double positive, DP) stage. The reduced levels of CD3 expression found at this stage of human thymocyte development raise the question of TCR (T-cell receptor) requirement for Foxp3 induction. We further show that, in mice, Foxp3 expression was also detected in pre-DP thymocytes of TCRalpha-sufficient but not in TCRalpha-deficient animals, genetically showing the TCR dependence of Foxp3 expression at pre-DP stages of T-cell development.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/biosynthesis , Receptors, Antigen, T-Cell/biosynthesis , T-Lymphocytes, Regulatory/cytology , Animals , CD4 Antigens/biosynthesis , CD4 Antigens/immunology , CD8 Antigens/biosynthesis , CD8 Antigens/immunology , Cell Lineage/immunology , Cell Separation , Flow Cytometry , Forkhead Transcription Factors/immunology , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
12.
Blood ; 113(13): 2999-3007, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19008454

ABSTRACT

The CD31(+) subset of human naive CD4(+) T cells is thought to contain the population of cells that have recently emigrated from the thymus, while their CD31(-) counterparts have been proposed to originate from CD31(+) cells after homeostatic cell division. Naive T-cell maintenance is known to involve homeostatic cytokines such as interleukin-7 (IL-7). It remains to be investigated what role this cytokine has in the homeostasis of naive CD4(+) T-cell subsets defined by CD31 expression. We provide evidence that IL-7 exerts a preferential proliferative effect on CD31(+) naive CD4(+) T cells from adult peripheral blood compared with the CD31(-) subset. IL-7-driven proliferation did not result in loss of CD31 expression, suggesting that CD31(+) naive CD4(+) T cells can undergo cytokine-driven homeostatic proliferation while preserving CD31. Furthermore, IL-7 sustained or increased CD31 expression even in nonproliferating cells. Both proliferation and CD31 maintenance were dependent on the activation of phosphoinositide 3-kinase (PI3K) signaling. Taken together, our data suggest that during adulthood CD31(+) naive CD4(+) T cells are maintained by IL-7 and that IL-7-based therapies may exert a preferential effect on this population.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Interleukin-7/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Adult , Aging/immunology , Aging/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Fetal Blood/cytology , Fetal Blood/immunology , Humans , Infant, Newborn , Interleukin-7/physiology , Organ Specificity/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/immunology , Signal Transduction/physiology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...