Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.128
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 41: 533-560, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36854182

ABSTRACT

Autoreactive B cells and interferons are central players in systemic lupus erythematosus (SLE) pathogenesis. The partial success of drugs targeting these pathways, however, supports heterogeneity in upstream mechanisms contributing to disease pathogenesis. In this review, we focus on recent insights from genetic and immune monitoring studies of patients that are refining our understanding of these basic mechanisms. Among them, novel mutations in genes affecting intrinsic B cell activation or clearance of interferogenic nucleic acids have been described. Mitochondria have emerged as relevant inducers and/or amplifiers of SLE pathogenesis through a variety of mechanisms that include disruption of organelle integrity or compartmentalization, defective metabolism, and failure of quality control measures. These result in extra- or intracellular release of interferogenic nucleic acids as well as in innate and/or adaptive immune cell activation. A variety of classic and novel SLE autoantibody specificities have been found to recapitulate genetic alterations associated with monogenic lupus or to trigger interferogenic amplification loops. Finally, atypical B cells and novel extrafollicular T helper cell subsets have been proposed to contribute to the generation of SLE autoantibodies. Overall, these novel insights provide opportunities to deepen the immunophenotypic surveillance of patients and open the door to patient stratification and personalized, rational approaches to therapy.


Subject(s)
Interferons , Lupus Erythematosus, Systemic , Humans , Animals , Interferons/therapeutic use , B-Lymphocytes , T-Lymphocytes, Helper-Inducer , Autoantibodies
2.
Annu Rev Immunol ; 41: 561-585, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37126418

ABSTRACT

Infection with SARS-CoV-2 results in clinical outcomes ranging from silent or benign infection in most individuals to critical pneumonia and death in a few. Genetic studies in patients have established that critical cases can result from inborn errors of TLR3- or TLR7-dependent type I interferon immunity, or from preexisting autoantibodies neutralizing primarily IFN-α and/or IFN-ω. These findings are consistent with virological studies showing that multiple SARS-CoV-2 proteins interfere with pathways of induction of, or response to, type I interferons. They are also congruent with cellular studies and mouse models that found that type I interferons can limit SARS-CoV-2 replication in vitro and in vivo, while their absence or diminution unleashes viral growth. Collectively, these findings point to insufficient type I interferon during the first days of infection as a general mechanism underlying critical COVID-19 pneumonia, with implications for treatment and directions for future research.


Subject(s)
COVID-19 , Interferon Type I , Mice , Humans , Animals , Interferons/pharmacology , SARS-CoV-2
3.
Annu Rev Immunol ; 36: 339-357, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29356584

ABSTRACT

Maintenance of immunological self-tolerance requires lymphocytes carrying self-reactive antigen receptors to be selectively prevented from mounting destructive or inflammatory effector responses. Classically, self-tolerance is viewed in terms of the removal, editing, or silencing of B and T cells that have formed self-reactive antigen receptors during their early development. However, B cells activated by foreign antigen can enter germinal centers (GCs), where they further modify their antigen receptor by somatic hypermutation (SHM) of their immunoglobulin genes. The inevitable emergence of activated, self-reactive GC B cells presents a unique challenge to the maintenance of self-tolerance that must be rapidly countered to avoid autoantibody production. Here we discuss current knowledge of the mechanisms that enforce B cell self-tolerance, with particular focus on the control of self-reactive GC B cells. We also consider how self-reactive GC B cells can escape self-tolerance to initiate autoantibody production or instead be redeemed via SHM and used in productive antibody responses.


Subject(s)
Autoimmunity , B-Lymphocytes/immunology , Germinal Center/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/metabolism , Germinal Center/metabolism , Humans , Immune Tolerance , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
4.
Annu Rev Immunol ; 34: 395-420, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26907212

ABSTRACT

Systemic autoimmune diseases are characterized by specific targeting of a limited group of ubiquitously expressed autoantigens by the immune system. This review examines the mechanisms underlying their selection as immune targets. Initiation of autoimmune responses likely reflects the presentation of antigens with a distinct structure not previously encountered by the immune system, in a proimmune context (injury, malignancy, or infection). Causes of modified structure include somatic mutation and posttranslational modifications (including citrullination and proteolysis). Many autoantigens are components of multimolecular complexes, and some of the other components may provide adjuvant activity. Propagation of autoimmune responses appears to reflect a bidirectional interaction between the immune response and the target tissues in a mutually reinforcing cycle: Immune effector pathways generate additional autoantigen, which feeds further immune response. We propose that this resonance may be a critical principle underlying disease propagation, with specific autoantigens functioning as the hubs around which amplification occurs.


Subject(s)
Autoantigens/immunology , Autoimmune Diseases/immunology , Immunodominant Epitopes/immunology , Multiprotein Complexes/immunology , Rheumatic Diseases/immunology , Adjuvants, Immunologic , Animals , Autoimmunity , Feedback, Physiological , Humans , Immune Tolerance , Structure-Activity Relationship
5.
Cell ; 185(7): 1208-1222.e21, 2022 03 31.
Article in English | MEDLINE | ID: mdl-35305314

ABSTRACT

The tumor microenvironment hosts antibody-secreting cells (ASCs) associated with a favorable prognosis in several types of cancer. Patient-derived antibodies have diagnostic and therapeutic potential; yet, it remains unclear how antibodies gain autoreactivity and target tumors. Here, we found that somatic hypermutations (SHMs) promote antibody antitumor reactivity against surface autoantigens in high-grade serous ovarian carcinoma (HGSOC). Patient-derived tumor cells were frequently coated with IgGs. Intratumoral ASCs in HGSOC were both mutated and clonally expanded and produced tumor-reactive antibodies that targeted MMP14, which is abundantly expressed on the tumor cell surface. The reversion of monoclonal antibodies to their germline configuration revealed two types of classes: one dependent on SHMs for tumor binding and a second with germline-encoded autoreactivity. Thus, tumor-reactive autoantibodies are either naturally occurring or evolve through an antigen-driven selection process. These findings highlight the origin and potential applicability of autoantibodies directed at surface antigens for tumor targeting in cancer patients.


Subject(s)
Antibodies, Neoplasm , Ovarian Neoplasms , Antibodies, Monoclonal , Autoantibodies , Autoantigens , Female , Humans , Ovarian Neoplasms/genetics , Tumor Microenvironment
6.
Cell ; 185(5): 881-895.e20, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35216672

ABSTRACT

Post-acute sequelae of COVID-19 (PASC) represent an emerging global crisis. However, quantifiable risk factors for PASC and their biological associations are poorly resolved. We executed a deep multi-omic, longitudinal investigation of 309 COVID-19 patients from initial diagnosis to convalescence (2-3 months later), integrated with clinical data and patient-reported symptoms. We resolved four PASC-anticipating risk factors at the time of initial COVID-19 diagnosis: type 2 diabetes, SARS-CoV-2 RNAemia, Epstein-Barr virus viremia, and specific auto-antibodies. In patients with gastrointestinal PASC, SARS-CoV-2-specific and CMV-specific CD8+ T cells exhibited unique dynamics during recovery from COVID-19. Analysis of symptom-associated immunological signatures revealed coordinated immunity polarization into four endotypes, exhibiting divergent acute severity and PASC. We find that immunological associations between PASC factors diminish over time, leading to distinct convalescent immune states. Detectability of most PASC factors at COVID-19 diagnosis emphasizes the importance of early disease measurements for understanding emergent chronic conditions and suggests PASC treatment strategies.


Subject(s)
COVID-19/complications , COVID-19/diagnosis , Convalescence , Adaptive Immunity/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Autoantibodies/blood , Biomarkers/metabolism , Blood Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Disease Progression , Female , Humans , Immunity, Innate/genetics , Longitudinal Studies , Male , Middle Aged , Risk Factors , SARS-CoV-2/isolation & purification , Transcriptome , Young Adult , Post-Acute COVID-19 Syndrome
7.
Cell ; 184(7): 1775-1789.e19, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33711260

ABSTRACT

Regulatory T cells prevent the emergence of autoantibodies and excessive IgE, but the precise mechanisms are unclear. Here, we show that BCL6-expressing Tregs, known as follicular regulatory T (Tfr) cells, produce abundant neuritin protein that targets B cells. Mice lacking Tfr cells or neuritin in Foxp3-expressing cells accumulated early plasma cells in germinal centers (GCs) and developed autoantibodies against histones and tissue-specific self-antigens. Upon immunization, these mice also produced increased plasma IgE and IgG1. We show that neuritin is taken up by B cells, causes phosphorylation of numerous proteins, and dampens IgE class switching. Neuritin reduced differentiation of mouse and human GC B cells into plasma cells, downregulated BLIMP-1, and upregulated BCL6. Administration of neuritin to Tfr-deficient mice prevented the accumulation of early plasma cells in GCs. Production of neuritin by Tfr cells emerges as a central mechanism to suppress B cell-driven autoimmunity and IgE-mediated allergies.


Subject(s)
B-Lymphocytes/immunology , Nerve Tissue Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoantibodies/immunology , Autoimmunity , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GPI-Linked Proteins/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Histones/immunology , Immunoglobulin Class Switching , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
8.
Cell ; 183(4): 968-981.e7, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32966765

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is typically very mild and often asymptomatic in children. A complication is the rare multisystem inflammatory syndrome in children (MIS-C) associated with COVID-19, presenting 4-6 weeks after infection as high fever, organ dysfunction, and strongly elevated markers of inflammation. The pathogenesis is unclear but has overlapping features with Kawasaki disease suggestive of vasculitis and a likely autoimmune etiology. We apply systems-level analyses of blood immune cells, cytokines, and autoantibodies in healthy children, children with Kawasaki disease enrolled prior to COVID-19, children infected with SARS-CoV-2, and children presenting with MIS-C. We find that the inflammatory response in MIS-C differs from the cytokine storm of severe acute COVID-19, shares several features with Kawasaki disease, but also differs from this condition with respect to T cell subsets, interleukin (IL)-17A, and biomarkers associated with arterial damage. Finally, autoantibody profiling suggests multiple autoantibodies that could be involved in the pathogenesis of MIS-C.


Subject(s)
Coronavirus Infections/pathology , Pneumonia, Viral/pathology , Systemic Inflammatory Response Syndrome/pathology , Autoantibodies/blood , Betacoronavirus/isolation & purification , COVID-19 , Child , Child, Preschool , Coronavirus Infections/complications , Coronavirus Infections/virology , Cytokines/metabolism , Female , Humans , Immunity, Humoral , Infant , Male , Mucocutaneous Lymph Node Syndrome/complications , Mucocutaneous Lymph Node Syndrome/immunology , Mucocutaneous Lymph Node Syndrome/pathology , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/virology , Principal Component Analysis , Proteome/analysis , SARS-CoV-2 , Severity of Illness Index , Systemic Inflammatory Response Syndrome/etiology , Systemic Inflammatory Response Syndrome/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Immunity ; 57(8): 1769-1779.e4, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38901428

ABSTRACT

Many infections, including malaria, are associated with an increase in autoantibodies (AAbs). Prior studies have reported an association between genetic markers of susceptibility to autoimmune disease and resistance to malaria, but the underlying mechanisms are unclear. Here, we performed a longitudinal study of children and adults (n = 602) in Mali and found that high levels of plasma AAbs before the malaria season independently predicted a reduced risk of clinical malaria in children during the ensuing malaria season. Baseline AAb seroprevalence increased with age and asymptomatic Plasmodium falciparum infection. We found that AAbs purified from the plasma of protected individuals inhibit the growth of blood-stage parasites and bind P. falciparum proteins that mediate parasite invasion. Protected individuals had higher plasma immunoglobulin G (IgG) reactivity against 33 of the 123 antigens assessed in an autoantigen microarray. This study provides evidence in support of the hypothesis that a propensity toward autoimmunity offers a survival advantage against malaria.


Subject(s)
Autoantibodies , Immunoglobulin G , Malaria, Falciparum , Plasmodium falciparum , Humans , Plasmodium falciparum/immunology , Autoantibodies/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Child , Child, Preschool , Adult , Immunoglobulin G/immunology , Immunoglobulin G/blood , Female , Mali , Male , Adolescent , Antibodies, Protozoan/immunology , Longitudinal Studies , Infant , Antigens, Protozoan/immunology , Young Adult , Autoantigens/immunology , Seroepidemiologic Studies , Middle Aged
10.
Immunity ; 57(7): 1457-1465, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38986441

ABSTRACT

Regardless of microbial virulence (i.e., the global infection-fatality ratio), age generally drives the prevalence of death from infection in unvaccinated humans. Four mortality patterns are recognized: the common U- and L-shaped curves of endemic infections and the unique W- and J-shaped curves of pandemic infections. We suggest that these patterns result from different sets of human genetic and immunological determinants. In this model, it is the interplay between (1) monogenic genotypes affecting immunity to primary infection that preferentially manifest early in life and related genotypes or their phenocopies, including auto-antibodies, which manifest later in life and (2) the occurrence and persistence of adaptive, acquired immunity to primary or cross-reactive infections, which shapes the age-dependent pattern of human deaths from infection.


Subject(s)
Communicable Diseases , Humans , Age Factors , Communicable Diseases/mortality , Communicable Diseases/immunology , Communicable Diseases/epidemiology , Adaptive Immunity/genetics , Aging/immunology , Aging/genetics , Pandemics
11.
Cell ; 170(5): 913-926.e19, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28841417

ABSTRACT

Germinal centers (GCs) are the primary sites of clonal B cell expansion and affinity maturation, directing the production of high-affinity antibodies. This response is a central driver of pathogenesis in autoimmune diseases, such as systemic lupus erythematosus (SLE), but the natural history of autoreactive GCs remains unclear. Here, we present a novel mouse model where the presence of a single autoreactive B cell clone drives the TLR7-dependent activation, expansion, and differentiation of other autoreactive B cells in spontaneous GCs. Once tolerance was broken for one self-antigen, autoreactive GCs generated B cells targeting other self-antigens. GCs became independent of the initial clone and evolved toward dominance of individual clonal lineages, indicating affinity maturation. This process produced serum autoantibodies to a breadth of self-antigens, leading to antibody deposition in the kidneys. Our data provide insight into the maturation of the self-reactive B cell response, contextualizing the epitope spreading observed in autoimmune disease.


Subject(s)
B-Lymphocytes/immunology , Clonal Evolution , Germinal Center/cytology , Germinal Center/immunology , Immune Tolerance , Animals , Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases/immunology , B-Lymphocytes/cytology , Chimera/immunology , Epitopes/immunology , Kidney/immunology , Mice , Mice, Inbred C57BL
12.
Proc Natl Acad Sci U S A ; 121(17): e2304199121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38630712

ABSTRACT

Although anti-citrullinated protein autoantibodies (ACPAs) are a hallmark serological feature of rheumatoid arthritis (RA), the mechanisms and cellular sources behind the generation of the RA citrullinome remain incompletely defined. Peptidylarginine deiminase IV (PAD4), one of the key enzymatic drivers of citrullination in the RA joint, is expressed by granulocytes and monocytes; however, the subcellular localization and contribution of monocyte-derived PAD4 to the generation of citrullinated autoantigens remain underexplored. In this study, we demonstrate that PAD4 displays a widespread cellular distribution in monocytes, including expression on the cell surface. Surface PAD4 was enzymatically active and capable of citrullinating extracellular fibrinogen and endogenous surface proteins in a calcium dose-dependent manner. Fibrinogen citrullinated by monocyte-surface PAD4 could be specifically recognized over native fibrinogen by a panel of eight human monoclonal ACPAs. Several unique PAD4 substrates were identified on the monocyte surface via mass spectrometry, with citrullination of the CD11b and CD18 components of the Mac-1 integrin complex being the most abundant. Citrullinated Mac-1 was found to be a target of ACPAs in 25% of RA patients, and Mac-1 ACPAs were significantly associated with HLA-DRB1 shared epitope alleles, higher C-reactive protein and IL-6 levels, and more erosive joint damage. Our findings implicate the monocyte cell surface as a unique and consequential site of extracellular and cell surface autoantigen generation in RA.


Subject(s)
Aminosalicylic Acids , Arthritis, Rheumatoid , Monocytes , Humans , Protein-Arginine Deiminases , Monocytes/metabolism , Autoantigens , Autoantibodies , Fibrinogen/metabolism , Citrulline/metabolism
13.
Immunol Rev ; 313(1): 181-193, 2023 01.
Article in English | MEDLINE | ID: mdl-36111456

ABSTRACT

The complement system plays an important pathophysiologic role in human diseases associated with immune or ischemic insults. In addition to understanding the effector mechanisms that are important for the biological effects of the system, substantial efforts have gone into understanding which specific complement activation pathways generate these potent effects. These approaches include the use of gene-targeted mice and specific pathway inhibitors, as well as the integration of human disease genetic and biomarker studies. In some disease states, it is quite clear that the alternate pathway plays a unique role in the initiation of the complement system. However, although initially a widely unexpected finding, it has now been shown in many tissue-based disease models and in initial human studies that engagement of the amplification loop is also essential for tissue injury when the classical and/or lectin pathways initiate pathway activation through pathogenic autoantibodies. This review provides evidence for such a conclusion through animal models, focusing on pathogenic antibody passive transfer models but also other relevant experimental systems. These data, along with initial biomarkers and clinical trial outcomes in human diseases that are associated with pathogenic autoantibodies, suggest that targeting the alternative pathway amplification loop may have near-universal therapeutic utility for tissue-based diseases.


Subject(s)
Complement Activation , Complement System Proteins , Humans , Animals , Mice , Disease Models, Animal , Biomarkers , Autoantibodies , Complement Pathway, Alternative
14.
Immunity ; 47(1): 107-117.e8, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28709804

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor Foxp3 are critical for the prevention of autoimmunity and the suppression of anti-tumor immunity. The major self-antigens recognized by Treg cells remain undefined, representing a substantial barrier to the understanding of immune regulation. Here, we have identified natural Treg cell ligands in mice. We found that two recurrent Treg cell clones, one prevalent in prostate tumors and the other associated with prostatic autoimmune lesions, recognized distinct non-overlapping MHC-class-II-restricted peptides derived from the same prostate-specific protein. Notably, this protein is frequently targeted by autoantibodies in experimental models of prostatic autoimmunity. On the basis of these findings, we propose a model in which Treg cell responses at peripheral sites converge on those self-proteins that are most susceptible to autoimmune attack, and we suggest that this link could be exploited as a generalizable strategy for identifying the Treg cell antigens relevant to human autoimmunity.


Subject(s)
Autoantigens/metabolism , Epitopes, T-Lymphocyte/metabolism , Prostatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/physiology , Animals , Autoantibodies/metabolism , Autoantigens/genetics , Autoantigens/immunology , Cell Differentiation , Clone Cells , Epitope Mapping , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation , Male , Mice
15.
Immunol Rev ; 307(1): 101-115, 2022 05.
Article in English | MEDLINE | ID: mdl-35001403

ABSTRACT

Autoimmune diseases are characterized by serum autoantibodies, some of which are pathogenic, causing severe manifestations and organ injury. However, autoantibodies of the same antigenic reactivity are also present in the serum of asymptomatic people years before they develop any clinical signs of autoimmunity. Autoantibodies can arise during multiple stages of B cell development, and various genetic and environmental factors drive their production. However, what drives the development of pathogenic autoantibodies is poorly understood. Advances in single-cell technology have enabled the deep analysis of rare B cell clones producing pathogenic autoantibodies responsible for vasculitis in patients with primary Sjögren's syndrome complicated by mixed cryoglobulinaemia. These findings demonstrated a cascade of genetic events involving stereotypic immunoglobulin V(D)J recombination and transforming somatic mutations in lymphoma genes and V(D)J regions that disrupted antibody quality control mechanisms and decreased autoantibody solubility. Most studies consider V(D)J mutations that enhance autoantibody affinity to drive pathology; however, V(D)J mutations that increase autoantibody propensity to form insoluble complexes could be a major contributor to autoantibody pathogenicity. Defining the molecular characteristics of pathogenic autoantibodies and failed tolerance checkpoints driving their formation will improve prognostication, enabling early treatment to prevent escalating organ damage and B cell malignancy.


Subject(s)
Autoantibodies , B-Lymphocytes , Autoimmunity/genetics , Clone Cells , Humans , Mutation/genetics
16.
Immunol Rev ; 307(1): 134-144, 2022 05.
Article in English | MEDLINE | ID: mdl-35092042

ABSTRACT

Better treatment of autoimmune diseases requires an improved understanding of the cellular and molecular mechanisms that lead to the breakdown of immune tolerance. The discovery of individuals with germline mutations in PIK3CD (which encodes the p110δ catalytic subunit of PI3K) has revealed the importance of regulated PI3Kδ activity to maintain tolerance. These patients display a range of symptoms including both immunodeficiency and autoimmunity. Here, we discuss recent advances in our understanding of how dysregulated PI3Kδ signaling affects the activation and differentiation of multiple cell types leading to the production of autoantibodies in these patients. This has lessons, not only for the treatment of these patients, but also for the potential role of dysregulated PI3Kδ in other patients with autoimmune conditions.


Subject(s)
Autoimmune Diseases , Immunologic Deficiency Syndromes , Autoimmune Diseases/genetics , Autoimmunity/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Immunologic Deficiency Syndromes/genetics , Signal Transduction/genetics
17.
Eur J Immunol ; 54(5): e2350682, 2024 May.
Article in English | MEDLINE | ID: mdl-38522030

ABSTRACT

SARS-CoV-2 typically causes mild symptoms in children, but evidence suggests that persistent immunopathological changes may lead to long COVID (LC). To explore the interplay between LC and innate immunity, we assessed the type I interferon (IFN-I) response in children and adolescents with LC symptoms (LC; n = 28). This was compared with age-matched SARS-CoV-2 recovered participants without LC symptoms (MC; n = 28) and healthy controls (HC; n = 18). We measured the mRNA expression of IFN-I (IFN-α/ß/ε/ω), IFN-I receptor (IFNAR1/2), and ISGs (ISG15, ISG56, MxA, IFI27, BST2, LY6E, OAS1, OAS2, OAS3, and MDA5) in PBMCs collected 3-6 months after COVID-19. LC adolescents (12-17 years) had higher transcript levels of IFN-ß, IFN-ε, and IFN-ω than HC, whereas LC children (6-11 years) had lower levels than HC. In adolescents, increased levels of IFN-α, IFN-ß, and IFN-ω mRNAs were found in the LC group compared with MC, while lower levels were observed in LC children than MC. Adolescents with neurological symptoms had higher IFN-α/ß mRNA levels than MC. LC and MC participants showed decreased expression of ISGs and IFNAR1, but increased expression of IFNAR2, than HC. Our results show age-related changes in the expression of transcripts involved in the IFN-I signaling pathway in children and adolescents with LC.


Subject(s)
COVID-19 , Interferon Type I , SARS-CoV-2 , Signal Transduction , Humans , Child , Adolescent , Interferon Type I/metabolism , Interferon Type I/immunology , Interferon Type I/genetics , Male , COVID-19/immunology , Female , Signal Transduction/immunology , SARS-CoV-2/immunology , Immunity, Innate , Age Factors , Post-Acute COVID-19 Syndrome , RNA, Messenger/genetics
18.
Brief Bioinform ; 25(1)2023 11 22.
Article in English | MEDLINE | ID: mdl-38243695

ABSTRACT

Idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of muscle disorders including adult and juvenile dermatomyositis, polymyositis, immune-mediated necrotising myopathy and sporadic inclusion body myositis, all of which present with variable symptoms and disease progression. The identification of effective biomarkers for IIMs has been challenging due to the heterogeneity between IIMs and within IIM subgroups, but recent advances in machine learning (ML) techniques have shown promises in identifying novel biomarkers. This paper reviews recent studies on potential biomarkers for IIM and evaluates their clinical utility. We also explore how data analytic tools and ML algorithms have been used to identify biomarkers, highlighting their potential to advance our understanding and diagnosis of IIM and improve patient outcomes. Overall, ML techniques have great potential to revolutionize biomarker discovery in IIMs and lead to more effective diagnosis and treatment.


Subject(s)
Autoimmune Diseases , Dermatomyositis , Myositis , Adult , Humans , Myositis/diagnosis , Myositis/therapy , Dermatomyositis/diagnosis , Biomarkers , Disease Progression
19.
FASEB J ; 38(3): e23460, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38315443

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death worldwide. There are currently no effective treatments for TBI, and trauma survivors suffer from a variety of long-lasting health consequences. With nutritional support recently emerging as a vital step in improving TBI patients' outcomes, we sought to evaluate the potential therapeutic benefits of nutritional supplements derived from bovine thymus gland, which can deliver a variety of nutrients and bioactive molecules. In a rat model of controlled cortical impact (CCI), we determined that animals supplemented with a nuclear fraction of bovine thymus (TNF) display greatly improved performance on beam balance and spatial memory tests following CCI. Using RNA-Seq, we identified an array of signaling pathways that are modulated by TNF supplementation in rat hippocampus, including those involved in the process of autophagy. We further show that bovine thymus-derived extracts contain antigens found in neural tissues and that supplementation of rats with thymus extracts induces production of serum IgG antibodies against neuronal and glial antigens, which may explain the enhanced animal recovery following CCI through possible oral tolerance mechanism. Collectively, our data demonstrate, for the first time, the potency of a nutritional supplement containing nuclear fraction of bovine thymus in enhancing the functional recovery from TBI.


Subject(s)
Brain Injuries, Traumatic , Thymus Extracts , Humans , Rats , Animals , Cattle , Thymus Extracts/pharmacology , Thymus Extracts/therapeutic use , Brain Injuries, Traumatic/drug therapy , Neurons , Neuroglia , Hippocampus , Disease Models, Animal
20.
Immunity ; 45(6): 1311-1326, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28002731

ABSTRACT

Liver X receptors (LXRs) are regulators of cholesterol metabolism that also modulate immune responses. Inactivation of LXR α and ß in mice leads to autoimmunity; however, how the regulation of cholesterol metabolism contributes to autoimmunity is unclear. Here we found that cholesterol loading of CD11c+ cells triggered the development of autoimmunity, whereas preventing excess lipid accumulation by promoting cholesterol efflux was therapeutic. LXRß-deficient mice crossed to the hyperlipidemic ApoE-deficient background or challenged with a high-cholesterol diet developed autoantibodies. Cholesterol accumulation in lymphoid organs promoted T cell priming and stimulated the production of the B cell growth factors Baff and April. Conversely, B cell expansion and the development of autoantibodies in ApoE/LXR-ß-deficient mice was reversed by ApoA-I expression. These findings implicate cholesterol imbalance as a contributor to immune dysfunction and suggest that stimulating HDL-dependent reverse cholesterol transport could be beneficial in the setting of autoimmune disease.


Subject(s)
Antigen-Presenting Cells/immunology , Autoimmune Diseases/immunology , Cholesterol/metabolism , Hypercholesterolemia/metabolism , Animals , Autoimmune Diseases/metabolism , CD11c Antigen/immunology , Cholesterol/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Hypercholesterolemia/immunology , Liver X Receptors/immunology , Liver X Receptors/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL