Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.146
Filter
1.
Front Immunol ; 15: 1373537, 2024.
Article in English | MEDLINE | ID: mdl-38812520

ABSTRACT

Sex-based differences in immune cell composition and function can contribute to distinct adaptive immune responses. Prior work has quantified these differences in peripheral blood, but little is known about sex differences within human lymphoid tissues. Here, we characterized the composition and phenotypes of adaptive immune cells from male and female ex vivo tonsils and evaluated their responses to influenza antigens using an immune organoid approach. In a pediatric cohort, female tonsils had more memory B cells compared to male tonsils direct ex vivo and after stimulation with live-attenuated but not inactivated vaccine, produced higher influenza-specific antibody responses. Sex biases were also observed in adult tonsils but were different from those measured in children. Analysis of peripheral blood immune cells from in vivo vaccinated adults also showed higher frequencies of tissue homing CD4 T cells in female participants. Together, our data demonstrate that distinct memory B and T cell profiles are present in male vs. female lymphoid tissues and peripheral blood respectively and suggest that these differences may in part explain sex biases in response to vaccines and viruses.


Subject(s)
Palatine Tonsil , Humans , Female , Male , Child , Palatine Tonsil/immunology , Adult , Influenza Vaccines/immunology , Influenza, Human/immunology , Sex Characteristics , Child, Preschool , Adolescent , Antibodies, Viral/blood , Antibodies, Viral/immunology , Memory B Cells/immunology , Organ Specificity/immunology , Young Adult , Sex Factors , CD4-Positive T-Lymphocytes/immunology , B-Lymphocytes/immunology , Immunologic Memory
2.
Nat Immunol ; 25(6): 1110-1122, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38698086

ABSTRACT

Lung-resident macrophages, which include alveolar macrophages and interstitial macrophages (IMs), exhibit a high degree of diversity, generally attributed to different activation states, and often complicated by the influx of monocytes into the pool of tissue-resident macrophages. To gain a deeper insight into the functional diversity of IMs, here we perform comprehensive transcriptional profiling of resident IMs and reveal ten distinct chemokine-expressing IM subsets at steady state and during inflammation. Similar IM subsets that exhibited coordinated chemokine signatures and differentially expressed genes were observed across various tissues and species, indicating conserved specialized functional roles. Other macrophage types shared specific IM chemokine profiles, while also presenting their own unique chemokine signatures. Depletion of CD206hi IMs in Pf4creR26EYFP+DTR and Pf4creR26EYFPCx3cr1DTR mice led to diminished inflammatory cell recruitment, reduced tertiary lymphoid structure formation and fewer germinal center B cells in models of allergen- and infection-driven inflammation. These observations highlight the specialized roles of IMs, defined by their coordinated chemokine production, in regulating immune cell influx and organizing tertiary lymphoid tissue architecture.


Subject(s)
Chemokines , Macrophages , Animals , Mice , Chemokines/metabolism , Macrophages/immunology , Macrophages/metabolism , Lung/immunology , Mice, Inbred C57BL , Inflammation/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Organ Specificity/immunology , Gene Expression Profiling , Mice, Transgenic , Tertiary Lymphoid Structures/immunology , Transcriptome
3.
Nature ; 629(8010): 174-183, 2024 May.
Article in English | MEDLINE | ID: mdl-38693412

ABSTRACT

Regular exercise promotes whole-body health and prevents disease, but the underlying molecular mechanisms are incompletely understood1-3. Here, the Molecular Transducers of Physical Activity Consortium4 profiled the temporal transcriptome, proteome, metabolome, lipidome, phosphoproteome, acetylproteome, ubiquitylproteome, epigenome and immunome in whole blood, plasma and 18 solid tissues in male and female Rattus norvegicus over eight weeks of endurance exercise training. The resulting data compendium encompasses 9,466 assays across 19 tissues, 25 molecular platforms and 4 training time points. Thousands of shared and tissue-specific molecular alterations were identified, with sex differences found in multiple tissues. Temporal multi-omic and multi-tissue analyses revealed expansive biological insights into the adaptive responses to endurance training, including widespread regulation of immune, metabolic, stress response and mitochondrial pathways. Many changes were relevant to human health, including non-alcoholic fatty liver disease, inflammatory bowel disease, cardiovascular health and tissue injury and recovery. The data and analyses presented in this study will serve as valuable resources for understanding and exploring the multi-tissue molecular effects of endurance training and are provided in a public repository ( https://motrpac-data.org/ ).


Subject(s)
Endurance Training , Multiomics , Physical Conditioning, Animal , Physical Endurance , Animals , Female , Humans , Male , Rats , Acetylation , Blood/immunology , Blood/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Databases, Factual , Epigenome , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Internet , Lipidomics , Metabolome , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Organ Specificity/physiology , Phosphorylation , Physical Conditioning, Animal/physiology , Physical Endurance/genetics , Physical Endurance/physiology , Proteome/metabolism , Proteomics , Time Factors , Transcriptome/genetics , Ubiquitination , Wounds and Injuries/genetics , Wounds and Injuries/immunology , Wounds and Injuries/metabolism
4.
Immunol Rev ; 323(1): 8-18, 2024 May.
Article in English | MEDLINE | ID: mdl-38628147

ABSTRACT

Cells of the mammalian innate immune system have evolved to protect the host from various environmental or internal insults and injuries which perturb the homeostatic state of the organism. Among the lymphocytes of the innate immune system are natural killer (NK) cells, which circulate and survey host tissues for signs of stress, including infection or transformation. NK cells rapidly eliminate damaged cells in the blood or within tissues through secretion of cytolytic machinery and production of proinflammatory cytokines. To perform these effector functions while traversing between the blood and tissues, patrolling NK cells require sufficient fuel to meet their energetic demands. Here, we highlight the ability of NK cells to metabolically adapt across tissues, during times of nutrient deprivation and within tumor microenvironments. Whether at steady state, or during viral infection and cancer, NK cells readily shift their nutrient uptake and usage in order to maintain metabolism, survival, and function.


Subject(s)
Killer Cells, Natural , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Immunity, Innate , Organ Specificity/immunology , Neoplasms/immunology , Neoplasms/metabolism , Energy Metabolism , Tumor Microenvironment/immunology , Homeostasis , Cytokines/metabolism , Metabolic Reprogramming
5.
Immunol Rev ; 323(1): 54-60, 2024 May.
Article in English | MEDLINE | ID: mdl-38568046

ABSTRACT

Natural killer (NK) cells are the prototype innate effector lymphocyte population that plays an important role in controlling viral infections and tumors. Studies demonstrating that NK cells form long-lived memory populations, akin to those generated by adaptive immune cells, prompted a revaluation of the potential functions of NK cells. Recent data demonstrating that NK cells are recruited from the circulation into tissues where they form long-lived memory-like populations further emphasize that NK cells have properties that mirror those of adaptive immune cells. NK cells that localize in non-lymphoid tissues are heterogeneous, and there is a growing appreciation that immune responses occurring within tissues are subject to tissue-specific regulation. Here we discuss both the immune effector and immunoregulatory functions of NK cells, with a particular emphasis on the role of NK cells within non-lymphoid tissues and how the tissue microenvironment shapes NK cell-dependent outcomes.


Subject(s)
Immunologic Memory , Killer Cells, Natural , Killer Cells, Natural/immunology , Humans , Animals , Organ Specificity/immunology , Adaptive Immunity , Cellular Microenvironment , Immunity, Innate
6.
J Immunol ; 208(4): 785-792, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35115374

ABSTRACT

Unlike the adaptive immune system, the innate immune system has classically been characterized as being devoid of memory functions. However, recent research shows that innate myeloid and lymphoid cells have the ability to retain memory of prior pathogen exposure and become primed to elicit a robust, broad-spectrum response to subsequent infection. This phenomenon has been termed innate immune memory or trained immunity. Innate immune memory is induced via activation of pattern recognition receptors and the actions of cytokines on hematopoietic progenitors and stem cells in bone marrow and innate leukocytes in the periphery. The trained phenotype is induced and sustained via epigenetic modifications that reprogram transcriptional patterns and metabolism. These modifications augment antimicrobial functions, such as leukocyte expansion, chemotaxis, phagocytosis, and microbial killing, to facilitate an augmented host response to infection. Alternatively, innate immune memory may contribute to the pathogenesis of chronic diseases, such as atherosclerosis and Alzheimer's disease.


Subject(s)
Communicable Diseases/etiology , Disease Susceptibility , Host-Pathogen Interactions/immunology , Immunity, Innate , Immunologic Memory , Animals , Biomarkers , Communicable Diseases/metabolism , Disease Resistance/genetics , Disease Resistance/immunology , Disease Susceptibility/immunology , Energy Metabolism , Epigenesis, Genetic , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction
7.
Molecules ; 27(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35209158

ABSTRACT

Dietary food components have the ability to affect immune function; following absorption, specifically orally ingested dietary food containing lectins can systemically modulate the immune cells and affect the response to self- and co-administered food antigens. The mannose-binding lectins from garlic (Allium sativum agglutinins; ASAs) were identified as immunodulatory proteins in vitro. The objective of the present study was to assess the immunogenicity and adjuvanticity of garlic agglutinins and to evaluate whether they have adjuvant properties in vivo for a weak antigen ovalbumin (OVA). Garlic lectins (ASA I and ASA II) were administered by intranasal (50 days duration) and intradermal (14 days duration) routes, and the anti-lectin and anti-OVA immune (IgG) responses in the control and test groups of the BALB/c mice were assessed for humoral immunogenicity. Lectins, co-administered with OVA, were examined for lectin-induced anti-OVA IgG response to assess their adjuvant properties. The splenic and thymic indices were evaluated as a measure of immunomodulatory functions. Intradermal administration of ASA I and ASA II had showed a four-fold and two-fold increase in anti-lectin IgG response, respectively, vs. the control on day 14. In the intranasal route, the increases were 3-fold and 2.4-fold for ASA I and ASA II, respectively, on day 50. No decrease in the body weights of animals was noticed; the increases in the spleen and thymus weights, as well as their indices, were significant in the lectin groups. In the adjuvanticity study by intranasal administration, ASA I co-administered with ovalbumin (OVA) induced a remarkable increase in anti-OVA IgG response (~six-fold; p < 0.001) compared to the control, and ASA II induced a four-fold increase vs. the control on day 50. The results indicated that ASA was a potent immunogen which induced mucosal immunogenicity to the antigens that were administered intranasally in BALB/c mice. The observations made of the in vivo study indicate that ASA I has the potential use as an oral and mucosal adjuvant to deliver candidate weak antigens. Further clinical studies in humans are required to confirm its applicability.


Subject(s)
Adjuvants, Immunologic , Garlic/chemistry , Immunity, Humoral , Lectins/immunology , Administration, Intranasal , Administration, Mucosal , Animals , Biomarkers , Enzyme-Linked Immunosorbent Assay , Immunization/methods , Immunoglobulin G/immunology , Immunomodulation , Lectins/administration & dosage , Lectins/isolation & purification , Mice , Mice, Inbred BALB C , Organ Specificity/immunology , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology
8.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055071

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by dysregulated T and B lymphocytes. Type I interferons (IFN-I) have been shown to play important pathogenic roles in both SLE patients and mouse models of lupus. Recent studies have shown that B cell intrinsic responses to IFN-I are enough to drive B cell differentiation into autoantibody-secreting memory B cells and plasma cells, although lower levels of residual auto-reactive cells remain present. We speculated that IFN-I stimulation of T cells would similarly drive specific T-cell associated lupus phenotypes including the upregulation of T follicular helper cells and Th17, thereby affecting autoantibody production and the development of glomerulonephritis. Using the B6.Nba2 mouse model of lupus, we evaluated disease parameters in T cell specific IFN-I receptor (IFNAR)-deficient mice (cKO). Surprisingly, all measured CD4+ T cell abnormalities and associated intra-splenic cytokine levels (IFNγ, IL-6, IL-10, IL-17, IL-21) were unchanged and thus independent of IFN-I. In contrast B6.Nba2 cKO mice displayed reduced levels of effector CD8+ T cells and increased levels of Foxp3+ CD8+ regulatory T cells, suggesting that IFN-I induced signaling specifically affecting CD8+ T cells. These data suggest a role for both pathogenic and immunosuppressive CD8+ T cells in Nba2-driven autoimmunity, providing a model to further evaluate the role of these cell subsets during lupus-like disease development in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Receptor, Interferon alpha-beta/metabolism , Animals , Antibodies, Antinuclear/immunology , Autoimmunity , Biomarkers , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cytokines , Disease Models, Animal , Germinal Center/immunology , Germinal Center/metabolism , Immunohistochemistry , Immunophenotyping , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Organ Specificity/immunology , Phenotype , Receptor, Interferon alpha-beta/genetics , Splenomegaly , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism
9.
Immunology ; 165(1): 3-21, 2022 01.
Article in English | MEDLINE | ID: mdl-34558663

ABSTRACT

An autoimmune disease is an inappropriate response to one's tissues due to a break in immune tolerance and exposure to self-antigens. It often leads to structural and functional damage to organs and systemic disorders. To date, there are no effective interventions to prevent the progression of autoimmune diseases. Hence, there is an urgent need for new treatment targets. TRPM7 is an enzyme-coupled, transient receptor ion channel of the subfamily M that plays a vital role in pathologic and physiologic conditions. While TRPM7 is constitutively activated under certain conditions, it can regulate cell migration, polarization, proliferation and cytokine secretion. However, a growing body of evidence highlights the critical role of TRPM7 in autoimmune diseases, including rheumatoid arthritis, multiple sclerosis and diabetes. Herein, we present (a) a review of the channel kinase properties of TRPM7 and its pharmacological properties, (b) discuss the role of TRPM7 in immune cells (neutrophils, macrophages, lymphocytes and mast cells) and its upstream immunoreactive substances, and (c) highlight TRPM7 as a potential therapeutic target for autoimmune diseases.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmunity , Immunomodulation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Biomarkers , Disease Susceptibility , Drug Development , Gene Expression Regulation/drug effects , Humans , Immune System/cytology , Immune System/drug effects , Immune System/immunology , Immune System/metabolism , Immunomodulation/drug effects , Ion Channel Gating/drug effects , Organ Specificity/drug effects , Organ Specificity/genetics , Organ Specificity/immunology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/chemistry , Signal Transduction/drug effects , Structure-Activity Relationship , TRPM Cation Channels/chemistry
10.
Immunology ; 165(1): 44-60, 2022 01.
Article in English | MEDLINE | ID: mdl-34716913

ABSTRACT

Cytokines are considered vital mediators of the immune system. Down- or upregulation of these mediators is linked to several inflammatory and pathologic situations. IL-26 is referred to as an identified member of the IL-10 family and IL-20 subfamily. Due to having a unique cationic structure, IL-26 exerts diverse functions in several diseases. Since IL-26 is mainly secreted from Th17, it is primarily considered a pro-inflammatory cytokine. Upon binding to its receptor complex (IL-10R1/IL-20R2), IL-26 activates multiple signalling mediators, especially STAT1/STAT3. In cancer, IL-26 induces IL-22-producing cells, which consequently decrease cytotoxic T-cell functions and promote tumour growth through activating anti-apoptotic proteins. In hypersensitivity conditions such as rheumatoid arthritis, multiple sclerosis, psoriasis and allergic disease, this cytokine functions primarily as the disease-promoting mediator and might be considered a biomarker for disease prognosis. Although IL-26 exerts antimicrobial function in infections such as hepatitis, tuberculosis and leprosy, it has also been shown that IL-26 might be involved in the pathogenesis and exacerbation of sepsis. Besides, the involvement of IL-26 has been confirmed in other conditions, including graft-versus-host disease and chronic obstructive pulmonary disease. Therefore, due to the multifarious function of this cytokine, it is proposed that the underlying mechanism regarding IL-26 function should be elucidated. Collectively, it is hoped that the examination of IL-26 in several contexts might be promising in predicting disease prognosis and might introduce novel approaches in the treatment of various diseases.


Subject(s)
Disease Susceptibility , Interleukins/genetics , Interleukins/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Infections/etiology , Infections/metabolism , Infections/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Interleukins/chemistry , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Protein Transport , Signal Transduction , Structure-Activity Relationship
11.
Life Sci Alliance ; 5(1)2022 01.
Article in English | MEDLINE | ID: mdl-34645668

ABSTRACT

Telomerase extends chromosome ends in somatic and germline stem cells to ensure continued proliferation. Mutations in genes critical for telomerase function result in telomeropathies such as dyskeratosis congenita, frequently resulting in spontaneous bone marrow failure. A dyskeratosis congenita mutation in TPP1 (K170∆) that specifically compromises telomerase recruitment to telomeres is a valuable tool to evaluate telomerase-dependent telomere length maintenance in mice. We used CRISPR-Cas9 to generate a mouse knocked in for the equivalent of the TPP1 K170∆ mutation (TPP1 K82∆) and investigated both its hematopoietic and germline compartments in unprecedented detail. TPP1 K82∆ caused progressive telomere erosion with increasing generation number but did not induce steady-state hematopoietic defects. Strikingly, K82∆ caused mouse infertility, consistent with gross morphological defects in the testis and sperm, the appearance of dysfunctional seminiferous tubules, and a decrease in germ cells. Intriguingly, both TPP1 K82∆ mice and previously characterized telomerase knockout mice show no spontaneous bone marrow failure but rather succumb to infertility at steady-state. We speculate that telomere length maintenance contributes differently to the evolutionary fitness of humans and mice.


Subject(s)
Dyskeratosis Congenita/diagnosis , Dyskeratosis Congenita/genetics , Germ Cells/metabolism , Hematopoiesis/genetics , Mutation , Telomere-Binding Proteins/genetics , Amino Acid Sequence , Animals , CRISPR-Cas Systems , Fertility/genetics , Gene Editing , Homozygote , Humans , Lymphopoiesis/genetics , Male , Mice , Mice, Knockout , Models, Molecular , Organ Specificity/genetics , Organ Specificity/immunology , Sperm Count , Structure-Activity Relationship
12.
Front Immunol ; 12: 803037, 2021.
Article in English | MEDLINE | ID: mdl-34970275

ABSTRACT

Macrophages are important immune cells in innate immunity, and have remarkable heterogeneity and polarization. Under pathological conditions, in addition to the resident macrophages, other macrophages are also recruited to the diseased tissues, and polarize to various phenotypes (mainly M1 and M2) under the stimulation of various factors in the microenvironment, thus playing different roles and functions. Liver diseases are hepatic pathological changes caused by a variety of pathogenic factors (viruses, alcohol, drugs, etc.), including acute liver injury, viral hepatitis, alcoholic liver disease, metabolic-associated fatty liver disease, liver fibrosis, and hepatocellular carcinoma. Recent studies have shown that macrophage polarization plays an important role in the initiation and development of liver diseases. However, because both macrophage polarization and the pathogenesis of liver diseases are complex, the role and mechanism of macrophage polarization in liver diseases need to be further clarified. Therefore, the origin of hepatic macrophages, and the phenotypes and mechanisms of macrophage polarization are reviewed first in this paper. It is found that macrophage polarization involves several molecular mechanisms, mainly including TLR4/NF-κB, JAK/STATs, TGF-ß/Smads, PPARγ, Notch, and miRNA signaling pathways. In addition, this paper also expounds the role and mechanism of macrophage polarization in various liver diseases, which aims to provide references for further research of macrophage polarization in liver diseases, contributing to the therapeutic strategy of ameliorating liver diseases by modulating macrophage polarization.


Subject(s)
Disease Susceptibility/immunology , Liver Diseases/etiology , Liver Diseases/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Plasticity/genetics , Cell Plasticity/immunology , Diagnosis, Differential , Energy Metabolism , Gene Expression Regulation , Humans , Liver Diseases/diagnosis , Liver Diseases/therapy , Macrophage Activation/genetics , Macrophages/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Phenotype , Signal Transduction
14.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638774

ABSTRACT

Targeted immunotherapies have greatly changed treatment of patients with B cell malignancies. To further enhance immunotherapies, research increasingly focuses on the tumor microenvironment (TME), which differs considerably by organ site. However, immunocompetent mouse models of disease to study immunotherapies targeting human molecules within organ-specific TME are surprisingly rare. We developed a myc-driven, primary murine lymphoma model expressing a human-mouse chimeric CD22 (h/mCD22). Stable engraftment of three distinct h/mCD22+ lymphoma was established after subcutaneous and systemic injection. However, only systemic lymphoma showed immune infiltration that reflected human disease. In this model, myeloid cells supported lymphoma growth and showed a phenotype of myeloid-derived suppressor cells. The human CD22-targeted immunotoxin Moxetumomab was highly active against h/mCD22+ lymphoma and similarly reduced infiltration of bone marrow and spleen of all three models up to 90-fold while efficacy against lymphoma in lymph nodes varied substantially, highlighting relevance of organ-specific TME. As in human aggressive lymphoma, anti-PD-L1 as monotherapy was not efficient. However, anti-PD-L1 enhanced efficacy of Moxetumomab suggesting potential for future clinical application. The novel model system of h/mCD22+ lymphoma provides a unique platform to test targeted immunotherapies and may be amenable for other human B cell targets such as CD19 and CD20.


Subject(s)
Immunotherapy , Lymphoma , Neoplasm Proteins , Neoplasms, Experimental , Sialic Acid Binding Ig-like Lectin 2 , Tumor Microenvironment , Animals , Humans , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/therapy , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Organ Specificity/genetics , Organ Specificity/immunology , Sialic Acid Binding Ig-like Lectin 2/genetics , Sialic Acid Binding Ig-like Lectin 2/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
15.
Int J Mol Sci ; 22(19)2021 Oct 03.
Article in English | MEDLINE | ID: mdl-34639062

ABSTRACT

The NOD, LRR, and pyrin domain-containing 3 (NLRP3) protein has been established as a central component of the inflammasome and regulates the inflammatory response to a myriad of environmental, microbial, and endogenous danger stimuli. Assembly of the NLRP3 inflammasome results in the cleavage and activation of caspase-1, in turn causing release of the pro-inflammatory interleukins 1-beta and 18. This activation response, while crucial to coordinated innate immune defense, can be aberrantly activated by the likes of cell-free DNA, and cause significant autoimmune pathology. Complications of autoimmunity induced by aberrant NLRP3 inflammasome activation have a great degree of mechanistic crossover with alloimmune injury in solid organ transplant, and stratagems to neutralize NLRP3 inflammasome activation may prove beneficial in solid organ transplant management. This article reviews NLRP3 inflammasome biology and the pathology associated with its hyperactivation, as well as the connections between NLRP3 inflammasome activation and allograft homeostasis.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Autoimmunity , DNA/immunology , Humans , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Organ Specificity/immunology , Organ Transplantation , Protein Processing, Post-Translational
16.
Sci Rep ; 11(1): 21110, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702961

ABSTRACT

Cryptococcus deneoformans is an opportunistic fungal pathogen that infects the lungs via airborne transmission and frequently causes fatal meningoencephalitis. Claudins (Cldns), a family of proteins with 27 members found in mammals, form the tight junctions within epithelial cell sheets. Cldn-4 and 18 are highly expressed in airway tissues, yet the roles of these claudins in respiratory infections have not been clarified. In the present study, we analyzed the roles of Cldn-4 and lung-specific Cldn-18 (luCldn-18) in host defense against C. deneoformans infection. luCldn-18-deficient mice exhibited increased susceptibility to pulmonary infection, while Cldn-4-deficient mice had normal fungal clearance. In luCldn-18-deficient mice, production of cytokines including IFN-γ was significantly decreased compared to wild-type mice, although infiltration of inflammatory cells including CD4+ T cells into the alveolar space was significantly increased. In addition, luCldn-18 deficiency led to high K+ ion concentrations in bronchoalveolar lavage fluids and also to alveolus acidification. The fungal replication was significantly enhanced both in acidic culture conditions and in the alveolar spaces of luCldn-18-deficient mice, compared with physiological pH conditions and those of wild-type mice, respectively. These results suggest that luCldn-18 may affect the clinical course of cryptococcal infection indirectly through dysregulation of the alveolar space microenvironment.


Subject(s)
Cellular Microenvironment/immunology , Claudins/deficiency , Cryptococcosis/immunology , Cryptococcus/immunology , Lung/immunology , Pneumonia/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cellular Microenvironment/genetics , Claudins/immunology , Cryptococcosis/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Lung/microbiology , Mice , Mice, Knockout , Organ Specificity/genetics , Organ Specificity/immunology , Pneumonia/genetics , Pneumonia/microbiology
17.
Sci Immunol ; 6(65): eabl9105, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34618554

ABSTRACT

Adaptive immune responses to SARS-CoV-2 infection have been extensively characterized in blood; however, most functions of protective immunity must be accomplished in tissues. Here, we report from examination of SARS-CoV-2 seropositive organ donors (ages 10 to 74) that CD4+ T, CD8+ T, and B cell memory generated in response to infection is present in the bone marrow, spleen, lung, and multiple lymph nodes (LNs) for up to 6 months after infection. Lungs and lung-associated LNs were the most prevalent sites for SARS-CoV-2­specific memory T and B cells with significant correlations between circulating and tissue-resident memory T and B cells in all sites. We further identified SARS-CoV-2­specific germinal centers in the lung-associated LNs up to 6 months after infection. SARS-CoV-2­specific follicular helper T cells were also abundant in lung-associated LNs and lungs. Together, the results indicate local tissue coordination of cellular and humoral immune memory against SARS-CoV-2 for site-specific protection against future infectious challenges.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Immunity, Cellular , Immunologic Memory , Lymphocytes/immunology , SARS-CoV-2/immunology , Female , Humans , Male , Organ Specificity/immunology
18.
Front Immunol ; 12: 703088, 2021.
Article in English | MEDLINE | ID: mdl-34557191

ABSTRACT

To effectively navigate complex tissue microenvironments, immune cells sense molecular concentration gradients using G-protein coupled receptors. However, due to the complexity of receptor activity, and the multimodal nature of chemokine gradients in vivo, chemokine receptor activity in situ is poorly understood. To address this issue, we apply a modelling and simulation approach that permits analysis of the spatiotemporal dynamics of CXCR5 expression within an in silico B-follicle with single-cell resolution. Using this approach, we show that that in silico B-cell scanning is robust to changes in receptor numbers and changes in individual kinetic rates of receptor activity, but sensitive to global perturbations where multiple parameters are altered simultaneously. Through multi-objective optimization analysis we find that the rapid modulation of CXCR5 activity through receptor binding, desensitization and recycling is required for optimal antigen scanning rates. From these analyses we predict that chemokine receptor signaling dynamics regulate migration in complex tissue microenvironments to a greater extent than the total numbers of receptors on the cell surface.


Subject(s)
B-Lymphocytes/immunology , Cellular Microenvironment/immunology , Models, Immunological , Receptors, CXCR5/immunology , Receptors, Chemokine/immunology , Signal Transduction/immunology , Humans , Organ Specificity/immunology
19.
Curr Opin Immunol ; 72: 331-339, 2021 10.
Article in English | MEDLINE | ID: mdl-34543865

ABSTRACT

The binary view of inborn errors of immunity classified as either autoinflammatory conditions or primary immunodeficiency in the strict sense, that is, increased susceptibility to infection is challenged by the description of recent inborn errors of immunity (IEI) triggers leading to activation and disruption of cell death pathways, play a major part in the pathophysiology of infection and autoinflammation. In addition, molecules with a double role in the extracellular versus intracellular milieu add to the complexity. In all, in-depth study of human inborn errors of immunity will continue to instruct us on fundamental immunology and lead to novel therapeutic targets and approaches that can be used in other monogenic and polygenic/complex immune disorders.


Subject(s)
Autoimmunity/genetics , Genetic Predisposition to Disease , Genetic Variation , Immunity/genetics , Infections/etiology , Inflammation/etiology , Alleles , Animals , Autoantibodies/immunology , Biomarkers , Endoplasmic Reticulum Stress , Genotype , Humans , Infections/diagnosis , Infections/metabolism , Inflammation/diagnosis , Inflammation/metabolism , Interferon Type I/immunology , Interferon Type I/metabolism , Mutation , Organ Specificity/genetics , Organ Specificity/immunology , Phenotype , Signal Transduction
20.
Front Immunol ; 12: 713540, 2021.
Article in English | MEDLINE | ID: mdl-34484216

ABSTRACT

Alternative splicing (AS) is a complex coordinated transcriptional regulatory mechanism. It affects nearly 95% of all protein-coding genes and occurs in nearly all human organs. Aberrant alternative splicing can lead to various neurological diseases and cancers and is responsible for aging, infection, inflammation, immune and metabolic disorders, and so on. Though aberrant alternative splicing events and their regulatory mechanisms are widely recognized, the association between autoimmune disease and alternative splicing has not been extensively examined. Autoimmune diseases are characterized by the loss of tolerance of the immune system towards self-antigens and organ-specific or systemic inflammation and subsequent tissue damage. In the present review, we summarized the most recent reports on splicing events that occur in the immunopathogenesis of systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) and attempted to clarify the role that splicing events play in regulating autoimmune disease progression. We also identified the changes that occur in splicing factor expression. The foregoing information might improve our understanding of autoimmune diseases and help develop new diagnostic and therapeutic tools for them.


Subject(s)
Alternative Splicing , Autoimmune Diseases/etiology , Gene Expression Regulation , Genetic Predisposition to Disease , Animals , Autoantibodies , Autoantigens/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmune Diseases/therapy , Autoimmunity/genetics , Biomarkers , Humans , Molecular Targeted Therapy , Mutation , Organ Specificity/genetics , Organ Specificity/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...