Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.454
Filter
1.
Immunity ; 47(1): 171-182.e4, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723549

ABSTRACT

Interleukin-7 (IL-7) availability determines the size and proliferative state of the resting T cell pool. However, the mechanisms that regulate steady-state IL-7 amounts are unclear. Using experimental lymphopenic mouse models and IL-7-induced homeostatic proliferation to measure IL-7 availability in vivo, we found that radioresistant cells were the source of IL-7 for both CD4+ and CD8+ T cells. Hematopoietic lineage cells, although irrelevant as a source of IL-7, were primarily responsible for limiting IL-7 availability via their expression of IL-7R. Unexpectedly, innate lymphoid cells were found to have a potent influence on IL-7 amounts in the primary and secondary lymphoid tissues. These results demonstrate that IL-7 homeostasis is achieved through consumption by multiple subsets of innate and adaptive immune cells.


Subject(s)
Hematopoietic Stem Cells/physiology , Interleukin-7/metabolism , Lymphocytes/immunology , Lymphopenia/immunology , T-Lymphocytes/physiology , Adaptive Immunity , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Homeostasis , Humans , Immunity, Innate , Interleukin-7/genetics , Interleukin-7/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Tolerance , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism
2.
Nat Immunol ; 14(2): 127-35, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263556

ABSTRACT

Interleukin 7 (IL-7) has a critical role in the development of early CD4(-)CD8(-) double-negative (DN) thymocytes. Although the transcription factor STAT5 is an important component of IL-7 signaling, differences in the phenotypes of mice deficient in STAT5, IL-7, IL-7 receptor alpha (IL-7rα) or the kinase Jak3 suggest the existence of STAT5-independent IL-7 signaling. Here we found that IL-7-Jak3 signals activated the transcription factor NFATc1 in DN thymocytes by phosphorylating Tyr371 in the regulatory region of NFATc1. This NFAT-activation pathway was critical for the survival and development of DN thymocytes, as deficiency in NFATc1 blocked thymocyte development at the DN1 stage, leading to T cell lymphopenia. In addition, our results demonstrated a cooperative function for NFATc1 and STAT5 in guiding thymocyte development in response to IL-7 signals.


Subject(s)
Interleukin-7/genetics , NFATC Transcription Factors/genetics , STAT5 Transcription Factor/genetics , Signal Transduction/immunology , Thymocytes/cytology , Animals , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Cell Differentiation , Gene Expression Regulation , Interleukin-7/immunology , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Transgenic , NFATC Transcription Factors/deficiency , Phosphorylation , Promoter Regions, Genetic , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , STAT5 Transcription Factor/immunology , Thymocytes/immunology , Transcription, Genetic
3.
Int Immunol ; 36(8): 413-424, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-38576231

ABSTRACT

Autoimmune diseases often arise from conditions where the immune system is compromised. While lymphopenia-induced proliferation (LIP) is crucial for immune system development and maturation, it is also caused by environmental insults, such as infection, and becomes a risk factor for autoimmunity in adults. We used Dsg3H1 TCR transgenic mice, whose T cells are designed to recognize desmogrein-3, a skin antigen, to explore the impact of lymphopenia on post-thymic tolerance. Dsg3H1 mice are known to delete the most highly autoreactive T cells in the thymus, and develop only subtle immune-mediated pathology in the steady state. However, we found that transient lymphopenia induced by total body irradiation (TBI) or cyclophosphamide (CY) results in massive dermatitis in Dsg3H1 mice. The symptoms included expansion and development of self-reactive T cells, their differentiation into CD44high IL-17-producing helper T cells, and severe neutrophilic inflammation. Repopulation of FOXP3+ T regulatory cells after lymphopenia normally occurred, suggesting escape of skin-reactive conventional T cells from control by regulatory T cells. Furthermore, we found that a depletion of the intestinal microbiota by antibiotics prevents CY-induced dermatitis, indicating roles of the commensal intestinal microbiota in LIP and Th17 development in vivo. The current data suggested that post-thymic tolerance of Dsg3H1 mice is established on a fragile balance in the lymphoreplete immune environment and broken by the interplay between lymphopenia and intestinal microbiota. The dynamic phenotypes observed in Dsg3H1 mice prompt a re-evaluation of opportunistic lymphopenia together with the microbiota as pivotal environmental factors, impacting individuals with genetic predispositions for autoimmune diseases.


Subject(s)
Gastrointestinal Microbiome , Immune Tolerance , Lymphopenia , Mice, Transgenic , Animals , Mice , Gastrointestinal Microbiome/immunology , Lymphopenia/immunology , Immune Tolerance/immunology , Skin/immunology , Skin/microbiology , Skin/pathology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/immunology , Mice, Inbred C57BL
4.
J Allergy Clin Immunol ; 154(3): 767-777, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38710235

ABSTRACT

BACKGROUND: LCP1 encodes L-plastin, an actin-bundling protein primarily expressed in hematopoietic cells. In mouse and fish models, LCP1 deficiency has been shown to result in hematologic and immune defects. OBJECTIVE: This study aimed to determine the nature of a human inborn error of immunity resulting from a novel genetic variant of LCP1. METHODS: We performed genetic, protein, and cellular analysis of PBMCs from a kindred with apparent autosomal dominant immune deficiency. We identified a candidate causal mutation in LCP1, which we evaluated by engineering the orthologous mutation in mice and Jurkat cells. RESULTS: A splice-site variant in LCP1 segregated with lymphopenia, neutropenia, and thrombocytopenia. The splicing defect resulted in at least 2 aberrant transcripts, producing an in-frame deletion of 24 nucleotides, and a frameshift deletion of exon 8. Cellular analysis of the kindred revealed a proportionate reduction of T and B cells and a mild expansion of transitional B cells. Similarly, mice carrying the orthologous genetic variant exhibited the same in-frame aberrant transcript, reduced expression Lcp1 and gene dose-dependent leukopenia, mild thrombocytopenia, and lymphopenia, with a significant reduction of T-cell populations. Functional analysis revealed that LCP1c740-1G>A confers a defect in platelet development and function with aberrant spreading on collagen. Immunologic analysis revealed defective actin organization in T cells, reduced migration of PBMCs from patients, splenocytes from mutant mice, and a mutant Jurkat cell line in response to CXCL12; impaired germinal center B-cell expansion after immunization; and reduced cytokinesis during T cell proliferation. CONCLUSIONS: We describe a unique human hematopoietic defect affecting neutrophils, lymphocytes, and platelets arising from partial LCP1 deficiency.


Subject(s)
Microfilament Proteins , Humans , Animals , Mice , Male , Female , Microfilament Proteins/genetics , Microfilament Proteins/deficiency , Thrombocytopenia/genetics , Thrombocytopenia/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphopenia/genetics , Lymphopenia/immunology , Jurkat Cells , Neutropenia/genetics , Neutropenia/immunology , Mutation , Pedigree , Cytopenia , Membrane Glycoproteins
5.
J Clin Immunol ; 45(1): 7, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264387

ABSTRACT

OBJECTIVE: To analyze the lymphocyte subsets in individuals with Kabuki syndrome for better characterizing the immunological phenotype of this rare congenital disorder. METHODS: We characterized the immunological profile including B-, T- and natural killer-cell subsets in a series (N = 18) of individuals with Kabuki syndrome. RESULTS: All 18 individuals underwent genetic analysis: 15 had a variant in KMT2D and 3 a variant in KDM6A. Eleven of the 18 individuals (61%) had recurrent infections and 9 (50%) respiratory infections. Three (17%) had autoimmune diseases. On immunological analysis, 6 (33%) had CD4 T-cell lymphopenia, which was preferentially associated with the KMT2D truncating variant (5/9 individuals). Eight of 18 individuals (44%) had a humoral deficiency and eight (44%) had B lymphopenia. We found abnormal distributions of T-cell subsets, especially a frequent decrease in recent thymic emigrant CD4 + naive T-cell count in 13/16 individuals (81%). CONCLUSION: The immunological features of Kabuki syndrome showed variable immune disorders with CD4 + T-cell deficiency in one third of cases, which had not been previously reported. In particular, we found a reduction in recent thymic emigrant naïve CD4 + T-cell count in 13 of 16 individuals, representing a novel finding that had not previously been reported.


Subject(s)
Abnormalities, Multiple , DNA-Binding Proteins , Face , Histone Demethylases , Neoplasm Proteins , Vestibular Diseases , Humans , Vestibular Diseases/genetics , Vestibular Diseases/immunology , Face/abnormalities , Female , Male , Abnormalities, Multiple/genetics , Abnormalities, Multiple/immunology , Child , DNA-Binding Proteins/genetics , Adolescent , Histone Demethylases/genetics , Child, Preschool , Adult , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Young Adult , Infant , Lymphopenia/immunology , Lymphopenia/genetics , Phenotype , Hematologic Diseases/genetics , Hematologic Diseases/immunology , Mutation , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Immunophenotyping
6.
Nat Immunol ; 13(7): 667-73, 2012 May 27.
Article in English | MEDLINE | ID: mdl-22634866

ABSTRACT

T cell-specific deletion of the receptor for transforming growth factor-ß (TGF-ß) mediated by Cre recombinase expressed early in T cell development leads to early-onset lethal autoimmune disease that cannot be controlled by regulatory T cells. However, when we deleted that receptor through the use of Cre driven by a promoter that is active much later in T cell development, adult mice in which most peripheral CD4(+) or CD8(+) T cells lacked the receptor for TGF-ß showed no signs of autoimmunity. Because of their enhanced responses to weak stimulation of the T cell antigen receptor, when transferred into lymphopenic recipients, naive TGF-ß-unresponsive T cells underwent much more proliferation and differentiation into effector cells and induced lymphoproliferative disease. We propose that TGF-ß signaling controls the self-reactivity of peripheral T cells but that in the absence of TGF-ß signals, an added trigger such as lymphopenia is needed to drive overt autoimmune disease.


Subject(s)
Autoimmunity/immunology , Cell Proliferation , Lymphopenia/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Animals , Cells, Cultured , Female , Leukocyte Common Antigens/immunology , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/immunology , Male , Mice , Receptors, Antigen, T-Cell/immunology , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology
7.
Cell Commun Signal ; 22(1): 349, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965547

ABSTRACT

T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.


Subject(s)
Basigin , COVID-19 , Lymphopenia , SARS-CoV-2 , Humans , Lymphopenia/immunology , Lymphopenia/virology , COVID-19/immunology , COVID-19/virology , COVID-19/pathology , SARS-CoV-2/metabolism , Basigin/metabolism , Angiotensin-Converting Enzyme 2/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Cytokine Release Syndrome/immunology , Animals
8.
J Immunol ; 208(3): 685-696, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34987111

ABSTRACT

Immune response dysregulation plays a key role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pathogenesis. In this study, we evaluated immune and endothelial blood cell profiles of patients with coronavirus disease 2019 (COVID-19) to determine critical differences between those with mild, moderate, or severe COVID-19 using spectral flow cytometry. We examined a suite of immune phenotypes, including monocytes, T cells, NK cells, B cells, endothelial cells, and neutrophils, alongside surface and intracellular markers of activation. Our results showed progressive lymphopenia and depletion of T cell subsets (CD3+, CD4+, and CD8+) in patients with severe disease and a significant increase in the CD56+CD14+Ki67+IFN-γ+ monocyte population in patients with moderate and severe COVID-19 that has not been previously described. Enhanced circulating endothelial cells (CD45-CD31+CD34+CD146+), circulating endothelial progenitors (CD45-CD31+CD34+/-CD146-), and neutrophils (CD11b+CD66b+) were coevaluated for COVID-19 severity. Spearman correlation analysis demonstrated the synergism among age, obesity, and hypertension with upregulated CD56+ monocytes, endothelial cells, and decreased T cells that lead to severe outcomes of SARS-CoV-2 infection. Circulating monocytes and endothelial cells may represent important cellular markers for monitoring postacute sequelae and impacts of SARS-CoV-2 infection during convalescence and for their role in immune host defense in high-risk adults after vaccination.


Subject(s)
COVID-19/immunology , Endothelial Cells/immunology , Monocytes/immunology , SARS-CoV-2 , Adolescent , Adult , Age Factors , Aged , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Biomarkers , CD56 Antigen/analysis , COVID-19/blood , COVID-19/epidemiology , Child , Comorbidity , Endothelial Cells/chemistry , Female , Flow Cytometry , Humans , Hypertension/epidemiology , Hypertension/immunology , Immunophenotyping , Lymphocyte Activation , Lymphocyte Subsets/immunology , Lymphopenia/etiology , Lymphopenia/immunology , Male , Middle Aged , Monocytes/chemistry , Neutrophils/immunology , Obesity/epidemiology , Obesity/immunology , Platelet Endothelial Cell Adhesion Molecule-1/analysis , SARS-CoV-2/immunology , Severity of Illness Index , Spike Glycoprotein, Coronavirus/immunology , Young Adult
9.
J Investig Allergol Clin Immunol ; 34(4): 233-245, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39077769

ABSTRACT

BACKGROUND: Common variable immunodeficiency (CVID) is considered the most symptomatic type of inborn errors of immunity in humans. Along with infectious complications, which have numerous consequences, noninfectious complications are a major challenge among CVID patients. METHODS: All CVID patients registered in the national database were included in this retrospective cohort study. Patients were divided into 2 groups based on the presence of B-cell lymphopenia. Demographic characteristics, laboratory findings, noninfectious organ involvement, autoimmunity, and lymphoproliferative diseases were evaluated. RESULTS: Among 387 enrolled patients, 66.4% were diagnosed with noninfectious complications and 33.6% with isolated infectious presentations. Enteropathy, autoimmunity, and lymphoproliferative disorders were reported in 35.1%, 24.3%, and 21.4% of patients, respectively. Some complications, including autoimmunity and hepatosplenomegaly, were reported to be significantly more frequent among patients with B-cell lymphopenia. As for organ involvement, the dermatologic, endocrine, and musculoskeletal systems were predominantly affected in CVID patients with B-cell lymphopenia. Among autoimmune manifestations, the frequency of rheumatologic, hematologic, and gastrointestinal autoimmunity was reported to be higher than that of other types of autoimmunity not associated with B cell-lymphopenia. Furthermore, hematological cancers, particularly lymphoma, were the most common type of malignancy. The mortality rate was 24.5%, and respiratory failure and malignancies were the most common causes of death, with no significant differences between the 2 groups. CONCLUSIONS: Considering that some of the noninfectious complications might be associated with B-cell lymphopenia, regular patient monitoring and follow-up with proper medication (in addition to immunoglobulin replacement therapy) are highly recommended to prevent sequelae and increase patient quality of life.


Subject(s)
B-Lymphocytes , Common Variable Immunodeficiency , Lymphopenia , Humans , Common Variable Immunodeficiency/complications , Common Variable Immunodeficiency/immunology , Female , Male , Adult , Retrospective Studies , B-Lymphocytes/immunology , Middle Aged , Lymphopenia/immunology , Young Adult , Autoimmunity , Adolescent , Aged , Child
10.
J Pak Med Assoc ; 74(5): 911-916, 2024 May.
Article in English | MEDLINE | ID: mdl-38783439

ABSTRACT

OBJECTIVE: To compare the extent of cytopenias and systemic immune inflammation index of hospitalised coronavirus disease-2019 patients during the first and second/third waves of the pandemic. Methods: The retrospective, cross-sectional study was conducted in October 2021 at Fatima Memorial Hospital, Lahore, Pakistan, and comprised data of hospitalised coronavirus disease-2019 patients regardless of age and gender from May 2020 to June 2021. Data was segregated into first wave that lasted from May to July 2020, second wave that lasted from early November to mid-December 2020, and third wave that ranged from mid-March to June 2021. For comparison purposes, the data of first wave was in group A, while data of second and third waves was pooled into group B. Age, gender, comorbidities, requirement of ventilator support and outcome of the patients was noted. Inflammatory markers were compared on the basis of complete blood count and systemic immune-inflammation index data. Data was analysed using SPSS 25. RESULTS: Of the 202 patients, 90(44.5%) were in group A and 112(55.4%) were in group B. There were 108(53.5%) males and 94(46.5%) females. The median age in males was 58 years (interquartile range: 21 years) and it was 56 years (interquartile range: 21 years) in females. Neutrophilia (p<0.001), leukocytosis (p<0.001) and lymphocytopenia (p<0.001) had direct association with increased systemic immune-inflammation. Raised systemic immune-inflammation also had an association with increased requirement of ventilator support (p=0.2) and increased mortality (p=0.001). There were more females, more critical patients, more patients with anaemia, leukopenia, lymphocytopenia and thrombocytopenia in group B compared to group A (p<0.05). Need for ventilator support and mortality were also higher in group B compared to group A (p<0.05). Conclusion: All the indicators analysed were worse during the second and third waves of coronavirus disease-2019 compared to the first wave of the pandemic.


Subject(s)
COVID-19 , SARS-CoV-2 , Thrombocytopenia , Humans , COVID-19/immunology , COVID-19/therapy , COVID-19/epidemiology , Male , Female , Middle Aged , Retrospective Studies , Cross-Sectional Studies , Pakistan/epidemiology , SARS-CoV-2/immunology , Adult , Aged , Thrombocytopenia/epidemiology , Thrombocytopenia/immunology , Hospitalization/statistics & numerical data , Leukopenia/epidemiology , Lymphopenia/immunology , Respiration, Artificial/statistics & numerical data , Inflammation/immunology , Cytopenia
SELECTION OF CITATIONS
SEARCH DETAIL