Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.632
2.
Immunol Rev ; 315(1): 108-125, 2023 05.
Article En | MEDLINE | ID: mdl-36653953

Historically, the immune system was believed to develop along a linear axis of maturity from fetal life to adulthood. Now, it is clear that distinct layers of immune cells are generated from unique waves of hematopoietic progenitors during different windows of development. This model, known as the layered immune model, has provided a useful framework for understanding why distinct lineages of B cells and γδ T cells arise in succession and display unique functions in adulthood. However, the layered immune model has not been applied to CD8+ T cells, which are still often viewed as a uniform population of cells belonging to the same lineage, with functional differences between cells arising from environmental factors encountered during infection. Recent studies have challenged this idea, demonstrating that not all CD8+ T cells are created equally and that the functions of individual CD8+ T cells in adults are linked to when they were created in the host. In this review, we discuss the accumulating evidence suggesting there are distinct ontogenetic subpopulations of CD8+ T cells and propose that the layered immune model be extended to the CD8+ T cell compartment.


CD8-Positive T-Lymphocytes , Immune System , T-Lymphocyte Subsets , Humans , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Human Development/physiology , Immune System/cytology , Immune System/growth & development , Immune System/immunology , Immune System/physiology , Immunity/immunology , Immunity/physiology , T-Lymphocyte Subsets/immunology
3.
Cells ; 11(14)2022 07 07.
Article En | MEDLINE | ID: mdl-35883580

Tumor suppressor WWOX inhibits cancer growth and retards Alzheimer's disease (AD) progression. Supporting evidence shows that the more strongly WWOX binds intracellular protein partners, the weaker is cancer cell growth in vivo. Whether this correlates with retardation of AD progression is unknown. Two functional forms of WWOX exhibit opposite functions. pY33-WWOX is proapoptotic and anticancer, and is essential for maintaining normal physiology. In contrast, pS14-WWOX is accumulated in the lesions of cancers and AD brains, and suppression of WWOX phosphorylation at S14 by a short peptide Zfra abolishes cancer growth and retardation of AD progression. In parallel, synthetic Zfra4-10 or WWOX7-21 peptide strengthens the binding of endogenous WWOX with intracellular protein partners leading to cancer suppression. Indeed, Zfra4-10 is potent in restoring memory loss in triple transgenic mice for AD (3xTg) by blocking the aggregation of amyloid beta 42 (Aß42), enhancing degradation of aggregated proteins, and inhibiting activation of inflammatory NF-κB. In light of the findings, Zfra4-10-mediated suppression of cancer and AD is due, in part, to an enhanced binding of endogenous WWOX and its binding partners. In this perspective review article, we detail the molecular action of WWOX in the HYAL-2/WWOX/SMAD4 signaling for biological effects, and discuss WWOX phosphorylation forms in interacting with binding partners, leading to suppression of cancer growth and retardation of AD progression.


Alzheimer Disease , Neoplasms , WW Domain-Containing Oxidoreductase , Adaptor Proteins, Signal Transducing/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Cell Survival , Disease Progression , Humans , Immunity/genetics , Immunity/physiology , Mice , Neoplasms/metabolism , Peptide Fragments/pharmacology , Protein Isoforms/metabolism , Tumor Suppressor Proteins/metabolism , WW Domain-Containing Oxidoreductase/metabolism
4.
Immunohorizons ; 6(7): 447-464, 2022 07 15.
Article En | MEDLINE | ID: mdl-35840326

Patients with STAT1 gain-of-function (GOF) pathogenic variants have enhanced or prolonged STAT1 phosphorylation following cytokine stimulation and exhibit increased yet heterogeneous susceptibility to infections, autoimmunity, and cancer. Although disease phenotypes are diverse and other genetic factors contribute, how STAT1 GOF affects cytokine sensitivity and cell biology remains poorly defined. In this study, we analyzed the immune and immunometabolic profiles of two patients with known pathogenic heterozygous STAT1 GOF mutation variants. A systems immunology approach of peripheral blood cells from these patients revealed major changes in multiple immune cell compartments relative to healthy adult and pediatric donors. Although many phenotypes of STAT1 GOF donors were shared, including increased Th1 cells but decreased class-switched B cells and plasmacytoid dendritic cell populations, others were heterogeneous. Mechanistically, hypersensitivity for cytokine-induced STAT1 phosphorylation in memory T cell populations was particularly evident in response to IL-6 in one STAT1 GOF patient. Immune cell metabolism directly influences cell function, and the STAT1 GOF patients shared an immunometabolic phenotype of heightened glucose transporter 1 (GLUT1) and carnitine palmitoyl transferase 1A (CPT1a) expression across multiple immune cell lineages. Interestingly, the metabolic phenotypes of the pediatric STAT1 GOF donors more closely resembled or exceeded those of healthy adult than healthy age-similar pediatric donors, which had low expression of these metabolic markers. These results define new features of STAT1 GOF patients, including a differential hypersensitivity for IL-6 and a shared increase in markers of metabolism in many immune cell types that suggests a role for STAT1 in metabolic regulation of immunity.


Immunity , STAT1 Transcription Factor , Cytokines/metabolism , Gain of Function Mutation/genetics , Humans , Immunity/genetics , Immunity/physiology , Interleukin-6 , Phenotype , Phosphorylation , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism
5.
Nat Commun ; 13(1): 2687, 2022 05 16.
Article En | MEDLINE | ID: mdl-35577777

Multisystem inflammatory syndrome in children (MIS-C) is an illness that emerged amidst the COVID-19 pandemic but shares many clinical features with the pre-pandemic syndrome of Kawasaki disease (KD). Here we compare the two syndromes using a computational toolbox of two gene signatures that were developed in the context of SARS-CoV-2 infection, i.e., the viral pandemic (ViP) and severe-ViP signatures and a 13-transcript signature previously demonstrated to be diagnostic for KD, and validated our findings in whole blood RNA sequences, serum cytokines, and formalin fixed heart tissues. Results show that KD and MIS-C are on the same continuum of the host immune response as COVID-19. Both the pediatric syndromes converge upon an IL15/IL15RA-centric cytokine storm, suggestive of shared proximal pathways of immunopathogenesis; however, they diverge in other laboratory parameters and cardiac phenotypes. The ViP signatures reveal unique targetable cytokine pathways in MIS-C, place MIS-C farther along in the spectrum in severity compared to KD and pinpoint key clinical (reduced cardiac function) and laboratory (thrombocytopenia and eosinopenia) parameters that can be useful to monitor severity.


COVID-19 , Mucocutaneous Lymph Node Syndrome , Systemic Inflammatory Response Syndrome , Artificial Intelligence , COVID-19/complications , COVID-19/genetics , COVID-19/immunology , Child , Computational Biology/methods , Cytokines , Gene Expression Profiling , Humans , Immunity/physiology , Mucocutaneous Lymph Node Syndrome/genetics , Mucocutaneous Lymph Node Syndrome/immunology , Pandemics , SARS-CoV-2 , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/immunology
6.
Cell Death Dis ; 13(2): 137, 2022 02 10.
Article En | MEDLINE | ID: mdl-35145069

Acute respiratory distress syndrome (ARDS) is triggered by various aetiological factors such as trauma, sepsis and respiratory viruses including SARS-CoV-2 and influenza A virus. Immune profiling of severe COVID-19 patients has identified a complex pattern of cytokines including granulocyte macrophage-colony stimulating factor (GM-CSF) and interleukin (IL)-5, which are significant mediators of viral-induced hyperinflammation. This strong response has prompted the development of therapies that block GM-CSF and other cytokines individually to limit inflammation related pathology. The common cytokine binding site of the human common beta (ßc) receptor signals for three inflammatory cytokines: GM-CSF, IL-5 and IL-3. In this study, ßc was targeted with the monoclonal antibody (mAb) CSL311 in engineered mice devoid of mouse ßc and ßIL-3 and expressing human ßc (hßcTg mice). Direct pulmonary administration of lipopolysaccharide (LPS) caused ARDS-like lung injury, and CSL311 markedly reduced lung inflammation and oedema, resulting in improved oxygen saturation levels in hßcTg mice. In a separate model, influenza (HKx31) lung infection caused viral pneumonia associated with a large influx of myeloid cells into the lungs of hßcTg mice. The therapeutic application of CSL311 potently decreased accumulation of monocytes/macrophages, neutrophils, and eosinophils without altering lung viral loads. Furthermore, CSL311 treatment did not limit the viral-induced expansion of NK and NKT cells, or the tissue expression of type I/II/III interferons needed for efficient viral clearance. Simultaneously blocking GM-CSF, IL-5 and IL-3 signalling with CSL311 may represent an improved and clinically applicable strategy to reducing hyperinflammation in the ARDS setting.


Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/physiology , Respiratory Distress Syndrome/immunology , Animals , Antibodies, Monoclonal/immunology , Cytokine Receptor Common beta Subunit/immunology , Cytokines , Eosinophils/immunology , Female , Humans , Immunity/genetics , Immunity/physiology , Inflammation/immunology , Leukocytes/metabolism , Male , Mice , Mice, Transgenic , Neutrophils/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Receptors, Interleukin-3 , Receptors, Interleukin-5 , Respiratory Distress Syndrome/physiopathology
7.
Int J Mol Sci ; 23(4)2022 Feb 15.
Article En | MEDLINE | ID: mdl-35216253

In recent years, several studies have demonstrated that polyunsaturated fatty acids have strong immunomodulatory properties, altering several functions of macrophages. In the present work, we sought to provide a multi-omic approach combining the analysis of the lipidome, the proteome, and the metabolome of RAW 264.7 macrophages supplemented with phospholipids containing omega-3 (PC 18:0/22:6; ω3-PC) or omega-6 (PC 18:0/20:4; ω6-PC) fatty acids, alone and in the presence of lipopolysaccharide (LPS). Supplementation of macrophages with ω3 and ω6 phospholipids plus LPS produced a significant reprogramming of the proteome of macrophages and amplified the immune response; it also promoted the expression of anti-inflammatory proteins (e.g., pleckstrin). Supplementation with the ω3-PC and ω6-PC induced significant changes in the lipidome, with a marked increase in lipid species linked to the inflammatory response, attributed to several pro-inflammatory signalling pathways (e.g., LPCs) but also to the pro-resolving effect of inflammation (e.g., PIs). Finally, the metabolomic analysis demonstrated that supplementation with ω3-PC and ω6-PC induced the expression of several metabolites with a pronounced inflammatory and anti-inflammatory effect (e.g., succinate). Overall, our data show that supplementation of macrophages with ω3-PC and ω6-PC effectively modulates the lipidome, proteome, and metabolome of these immune cells, affecting several metabolic pathways involved in the immune response that are triggered by inflammation.


Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6/metabolism , Immunologic Factors/metabolism , Lipids/physiology , Macrophages/metabolism , Phospholipids/metabolism , Proteins/metabolism , Animals , Immunity/physiology , Inflammation/metabolism , Lipidomics/methods , Metabolome/physiology , Mice , Proteome/metabolism , RAW 264.7 Cells , Signal Transduction/physiology
8.
Cell Rep ; 38(3): 110271, 2022 01 18.
Article En | MEDLINE | ID: mdl-35026155

The utility of the urinary proteome in infectious diseases remains unclear. Here, we analyzed the proteome and metabolome of urine and serum samples from patients with COVID-19 and healthy controls. Our data show that urinary proteins effectively classify COVID-19 by severity. We detect 197 cytokines and their receptors in urine, but only 124 in serum using TMT-based proteomics. The decrease in urinary ESCRT complex proteins correlates with active SARS-CoV-2 replication. The downregulation of urinary CXCL14 in severe COVID-19 cases positively correlates with blood lymphocyte counts. Integrative multiomics analysis suggests that innate immune activation and inflammation triggered renal injuries in patients with COVID-19. COVID-19-associated modulation of the urinary proteome offers unique insights into the pathogenesis of this disease. This study demonstrates the added value of including the urinary proteome in a suite of multiomics analytes in evaluating the immune pathobiology and clinical course of COVID-19 and, potentially, other infectious diseases.


COVID-19/urine , Immunity , Metabolome , Proteome/analysis , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/blood , COVID-19/immunology , COVID-19/pathology , Case-Control Studies , Child , Child, Preschool , China , Cohort Studies , Female , Humans , Immunity/physiology , Male , Metabolome/immunology , Metabolomics , Middle Aged , Patient Acuity , Proteome/immunology , Proteome/metabolism , Proteomics , Urinalysis/methods , Young Adult
9.
Int J Biol Sci ; 18(2): 617-636, 2022.
Article En | MEDLINE | ID: mdl-35002513

Among numerous studies on coronavirus 2019 (COVID-19), we noted that the infection and mortality rates of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) increased with age and that fetuses known to be particularly susceptible to infection were better protected despite various mutations. Hence, we established the hypothesis that a new immune system exists that forms before birth and decreases with aging. Methods: To prove this hypothesis, we established new ex-vivo culture conditions simulating the critical environmental factors of fetal stem cells (FSCs) in early pregnancy. Then, we analyzed the components from FSCs cultivated newly developed ex-vivo culture conditions and compared them from FSCs cultured in a normal condition. Results: We demonstrated that immunoglobulin M (IgM), a natural antibody (NAb) produced only in early B-1 cells, immunoglobulins (Igs) including IgG3, which has a wide range of antigen-binding capacity and affinity, complement proteins, and antiviral proteins are induced in FSCs only cultured in newly developed ex-vivo culture conditions. Particularly we confirmed that their extracellular vesicles (EVs) contained NAbs, Igs, various complement proteins, and antiviral proteins, as well as human leukocyte antigen G (HLA-G), responsible for immune tolerance. Conclusion: Our results suggest that FSCs in early pregnancy can form an independent immune system responding to unlearned antigens as a self-defense mechanism before establishing mature immune systems. Moreover, we propose the possibility of new solutions to cope with various infectious diseases based on the factors in NAbs-containing EVs, especially not causing unnecessary immune reaction due to HLA-G.


Aging/immunology , COVID-19/immunology , Fetal Stem Cells/physiology , Immunity/physiology , SARS-CoV-2 , Antibody Affinity , Antigens, Viral , Antiglaucoma Agents , Complement System Proteins , Female , Fetal Stem Cells/virology , Gene Expression Regulation, Developmental/immunology , Humans , Immunoglobulins/metabolism , Pregnancy
10.
Curr Opin Insect Sci ; 50: 100874, 2022 04.
Article En | MEDLINE | ID: mdl-35051619

Activation of an immune response is energetically costly and excessive immune system activity can result in immunopathology, yet a slow or insufficient immune response carries the risk of pathogen establishment with consequent pathology arising from the infection. Mathematical theory and empirical data demonstrate that hosts balance the costs of immunity against the risk of infection by closely regulating immunological dynamics. An optimal immune system is rapidly and robustly deployed against a true infectious threat and rapidly deactivated once the threat has been controlled. Genetic variation in the sensitivity of an immune system, as well as in the activation and shutdown kinetics of host immune responses, can contribute to the evolution of pathogen virulence and host tolerance of infection. Improved understanding of the adaptive forces that operate on immune regulatory dynamics will clarify fundamental principles governing the evolution and maintenance of innate immune systems.


Immunity , Virulence , Animals , Immunity/physiology , Risk
11.
Cell Rep ; 38(2): 110090, 2022 01 11.
Article En | MEDLINE | ID: mdl-35021075

Alpha-synuclein (αS) is causally involved in the development of Parkinson disease (PD); however, its role in normal vertebrate physiology has remained unknown. Recent studies demonstrate that αS is induced by noroviral infection in the enteric nervous system of children and protects mice against lethal neurotropic viral infection. Additionally, αS is a potent chemotactic activator of phagocytes. In this report, using both wild-type and αS knockout mice, we show that αS is a critical mediator of inflammatory and immune responses. αS is required for the development of a normal inflammatory response to bacterial peptidoglycan introduced into the peritoneal cavity as well as antigen-specific and T cell responses following intraperitoneal immunization. Furthermore, we show that neural cells are the sources of αS required for immune competence. Our report supports the hypothesis that αS accumulates within the nervous system of PD individuals because of an inflammatory/immune response.


Immunity/physiology , alpha-Synuclein/metabolism , alpha-Synuclein/physiology , Animals , Brain/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System/metabolism , Neurons/metabolism , Toll-Like Receptor 4/immunology , alpha-Synuclein/genetics
13.
Gastroenterology ; 162(2): 548-561.e4, 2022 02.
Article En | MEDLINE | ID: mdl-34687739

BACKGROUND AND AIMS: Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with altered gut microbiota composition. Phylogenetic groups of gut bacteria involved in the metabolism of short chain fatty acids (SCFAs) were depleted in SARS-CoV-2-infected patients. We aimed to characterize a functional profile of the gut microbiome in patients with COVID-19 before and after disease resolution. METHODS: We performed shotgun metagenomic sequencing on fecal samples from 66 antibiotics-naïve patients with COVID-19 and 70 non-COVID-19 controls. Serial fecal samples were collected (at up to 6 times points) during hospitalization and beyond 1 month after discharge. We assessed gut microbial pathways in association with disease severity and blood inflammatory markers. We also determined changes of microbial functions in fecal samples before and after disease resolution and validated these functions using targeted analysis of fecal metabolites. RESULTS: Compared with non-COVID-19 controls, patients with COVID-19 with severe/critical illness showed significant alterations in gut microbiome functionality (P < .001), characterized by impaired capacity of gut microbiome for SCFA and L-isoleucine biosynthesis and enhanced capacity for urea production. Impaired SCFA and L-isoleucine biosynthesis in gut microbiome persisted beyond 30 days after recovery in patients with COVID-19. Targeted analysis of fecal metabolites showed significantly lower fecal concentrations of SCFAs and L-isoleucine in patients with COVID-19 before and after disease resolution. Lack of SCFA and L-isoleucine biosynthesis significantly correlated with disease severity and increased plasma concentrations of CXCL-10, NT- proB-type natriuretic peptide, and C-reactive protein (all P < .05). CONCLUSIONS: Gut microbiome of patients with COVID-19 displayed impaired capacity for SCFA and L-isoleucine biosynthesis that persisted even after disease resolution. These 2 microbial functions correlated with host immune response underscoring the importance of gut microbial functions in SARS-CoV-2 infection pathogenesis and outcome.


COVID-19/microbiology , Fatty Acids, Volatile/biosynthesis , Gastrointestinal Microbiome/genetics , Immunity/physiology , Isoleucine/biosynthesis , Adult , Biomarkers/blood , Case-Control Studies , Feces/microbiology , Female , Humans , Male , Metagenomics , Middle Aged , Phylogeny , SARS-CoV-2 , Severity of Illness Index
14.
Med Sci Sports Exerc ; 54(3): 377-387, 2022 03 01.
Article En | MEDLINE | ID: mdl-34690285

PURPOSE: To quantify the effects of a short-term (6-d) low carbohydrate (CHO) high fat (LCHF), and low energy availability (LEA) diet on immune, inflammatory, and iron-regulatory responses to exercise in endurance athletes. METHODS: Twenty-eight elite male race walkers completed two 6-d diet/training phases. During phase 1 (Baseline), all athletes consumed a high CHO/energy availability (CON) diet (65% CHO and ~40 kcal·kg-1 fat-free mass (FFM)·d-1). In phase 2 (Adaptation), athletes were allocated to either a CON (n = 10), LCHF (n = 8; <50 g·d-1 CHO and ~40 kcal·kg-1·FFM-1·d-1), or LEA diet (n = 10; 60% CHO and 15 kcal·kg-1·FFM-1·d-1). At the end of each phase, athletes completed a 25-km race walk protocol at ~75% V˙O2max. On each occasion, venous blood was collected before and after exercise for interleukin-6, hepcidin, cortisol, and glucose concentrations, as well as white blood cell counts. RESULTS: The LCHF athletes displayed a greater IL-6 (P = 0.019) and hepcidin (P = 0.011) response to exercise after Adaptation, compared with Baseline. Similarly, postexercise increases in total white blood cell counts (P = 0.026) and cortisol levels (P < 0.001) were larger compared with Baseline after LCHF Adaptation. Decreases in blood glucose concentrations were evident postexercise during Adaptation in LCHF (P = 0.049), whereas no change occurred in CON or LEA (P > 0.05). No differences between CON and LEA were evident for any of the measured biological markers (all P > 0.05). CONCLUSIONS: Short-term adherence to a LCHF diet elicited small yet unfavorable iron, immune, and stress responses to exercise. In contrast, no substantial alterations to athlete health were observed when athletes restricted energy availability compared with athletes with adequate energy availability. Therefore, short-term restriction of CHO, rather than energy, may have greater negative impacts on athlete health.


Athletes , Athletic Performance/physiology , Diet, Ketogenic/methods , Immunity/physiology , Iron/metabolism , Walking/physiology , Adult , Biomarkers/blood , Humans , Male , Young Adult
15.
Immunol Invest ; 51(5): 1347-1363, 2022 Jul.
Article En | MEDLINE | ID: mdl-34121590

BACKGROUND: Leptin plays an important role in the regulation of the immune response. There is a physiological surge of leptin in rodents during the neonatal period, which has mainly been studied in the context of brain development. However, little is known about the effects of this neonatal leptin surge on immunity. Therefore, we investigated whether blocking this leptin surge could affect several immune functions. METHODS: Male and female rats were injected subcutaneously with 5 mg/Kg/day of rat pegylated super leptin antagonist during the neonatal period (PND5-9). On the peripubertal period, relevant functions as well as cytokine release by spleen leukocytes were studied in these animals. RESULTS: The results showed that the animals significantly display an impaired anti-tumor NK activity and chemotactic and proliferation capacity of lymphocytes in response to mitogens. In addition, several cytokine concentrations, released under mitogen-stimulated conditions, were also altered. CONCLUSION: In conclusion, the neonatal leptin surge seems to be involved in the establishment of an adequate immune response and cytokine profile, which are crucial for the maintenance of a healthy life.


Growth and Development , Leptin , Animals , Animals, Newborn/growth & development , Animals, Newborn/immunology , Cytokines/analysis , Cytokines/immunology , Female , Growth and Development/immunology , Immunity/immunology , Immunity/physiology , Intercellular Signaling Peptides and Proteins/immunology , Leptin/immunology , Male , Rats/immunology
16.
Article En | MEDLINE | ID: mdl-33583391

Angiotensin-converting enzyme (ACE) is a zinc-dependent dicarboxypeptidase with two catalytic components, which has an important role in regulating blood pressure by converting angiotensin I to angiotensin II. ACE breaks down other peptides besides angiotensin I and has a variety of physiological effects together with renal growth and reproduction in men. ACE also acts on innate and acquired immune systems by affecting macrophage and neutrophil function, and these outcomes are exacerbated due to the overexpression of ACE. Overexpression of ACE in macrophages imposes antitumor and antimicrobial response, and it enhances the ability of neutrophils to produced super peroxide that has a bactericidal effect. ACE is also known to contribute to the expression of Major Histocompatibility Complex (MHC) class I and MHC class II peptides through enzymatic alterations of these peptides. Apprehending the expression of ACE and its effects on myeloid cell (myelogenous cells) activity can be promising in therapeutic interventions, including treatment of infection and malignancy.


Immunity/genetics , Peptidyl-Dipeptidase A/physiology , Angiotensins/metabolism , Angiotensins/physiology , Animals , Antigen Presentation/genetics , Antigen Presentation/physiology , Female , Histocompatibility Antigens Class I/physiology , Histocompatibility Antigens Class II/physiology , Humans , Immunity/physiology , Infections/genetics , Infections/immunology , Infections/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Peptidyl-Dipeptidase A/genetics
17.
Parasite Immunol ; 44(3): e12903, 2022 03.
Article En | MEDLINE | ID: mdl-34964129

Circadian rhythms are recurring variations of physiology with a period of ~24 h, generated by circadian clocks located throughout the body. Studies have shown a circadian regulation of many aspects of immunity. Immune cells have intrinsic clock mechanisms, and innate and adaptive immune responses - such as leukocyte migration, magnitude of inflammation, cytokine production and cell differentiation - are under circadian control. This circadian regulation has consequences for infections including parasitic infections. In the context of Leishmania infection, the circadian clock within host immune cells modulates the magnitude of the infection and the inflammatory response triggered by the parasite. As for malaria, rhythms within the immune system were shown to impact the developmental cycles of Plasmodium parasites within red blood cells. Further, host circadian rhythms impact infections by multicellular parasites; for example, infection with helminth Trichuris muris shows different kinetics of worm expulsion depending on time of day of infection, a variation that depends on the dendritic cell clock. Although the research on the circadian control of immunity in the context of parasitic infections is in its infancy, the research reviewed here suggests a crucial involvement of host circadian rhythms in immunity on the development and progression of parasitic infections.


Circadian Clocks , Parasitic Diseases , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Immunity/physiology , Mammals
18.
Neuropeptides ; 91: 102213, 2022 Feb.
Article En | MEDLINE | ID: mdl-34839164

Pregnancy, from insemination to parturition, is a highly complex but well-orchestrated process that requires various organs and systems to participate. Immune system and neuroendocrine system are important regulators in healthy pregnancy. Dozens of neuroactive factors have been detected in human placenta, whether they are locally secreted or circulated. Among them, some are vividly studied such as corticotropin-releasing hormone (CRH), human chorionic gonadotropin (hCG), transforming growth factor-ß (TGF-ß), progesterone and estrogens, while others are relatively lack of research. Though the neuroendocrine-immune interactions are demonstrated in some diseases for decades, the roles of neuroactive factors in immune system and lymphocytes during pregnancy are not fully elucidated. This review aims to provide an interdisciplinary view on how the neuroendocrine system mediate immune system during pregnancy process.


Brain/immunology , Immunity/physiology , Placenta/immunology , Animals , Female , Humans , Pregnancy
19.
Cell Rep ; 37(11): 110112, 2021 12 14.
Article En | MEDLINE | ID: mdl-34863353

An ideal vaccine against SARS-CoV-2 is expected to elicit broad immunity to prevent viral infection and disease, with efficient viral clearance in the upper respiratory tract (URT). Here, the N protein and prefusion-full S protein (SFLmut) are combined with flagellin (KF) and cyclic GMP-AMP (cGAMP) to generate a candidate vaccine, and this vaccine elicits stronger systemic and mucosal humoral immunity than vaccines containing other forms of the S protein. Furthermore, the candidate vaccine administered via intranasal route can enhance local immune responses in the respiratory tract. Importantly, human ACE2 transgenic mice given the candidate vaccine are protected against lethal SARS-CoV-2 challenge, with superior protection in the URT compared with that in mice immunized with an inactivated vaccine. In summary, the developed vaccine can elicit a multifaceted immune response and induce robust viral clearance in the URT, which makes it a potential vaccine for preventing disease and infection of SARS-CoV-2.


COVID-19 Vaccines/immunology , Combined Modality Therapy/methods , SARS-CoV-2/immunology , Adjuvants, Vaccine , Administration, Intranasal , Angiotensin-Converting Enzyme 2/immunology , Angiotensin-Converting Enzyme 2/metabolism , Animals , Antibodies, Viral/immunology , Antigens/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/genetics , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/immunology , Female , Flagellin/immunology , HEK293 Cells , Humans , Immunity/immunology , Immunity/physiology , Immunity, Humoral/immunology , Immunization , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nucleotides, Cyclic/immunology , Phosphoproteins/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/immunology , Vaccination , Vero Cells
20.
Genes (Basel) ; 12(11)2021 10 26.
Article En | MEDLINE | ID: mdl-34828304

Beyond its role on the conversion of nutrients into energy and biomass, cellular metabolism is actively involved in the control of many physiological processes. Among these, it is becoming increasingly evident that specific metabolic pathways are associated with the phenotype of several immune cell types and, importantly, are crucial in controlling their differentiation, proliferation, and effector functions, thus shaping the immune response against pathogens and tumors. In this context, data generated over the last decade have uncovered mammalian sirtuins as important regulators of cellular metabolism, immune cell function, and cancer. Here, we summarize our current knowledge on the roles of this family of protein deacylases on the metabolic control of immune cells and their implications on immune-related diseases and cancer.


Immune System/metabolism , Immunity/physiology , Sirtuins/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Energy Metabolism/genetics , Humans , Immune System/cytology , Immunity/genetics , Mitochondria/metabolism , Phenotype
...