Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.492
Filter
1.
Proc Natl Acad Sci U S A ; 121(39): e2316161121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39298490

ABSTRACT

Uveitis is a vision-threatening disease primarily driven by a dysregulated immune response, with retinal microglia playing a pivotal role in its progression. Although the transcription factor EGR2 is known to be closely associated with uveitis, including Vogt-Koyanagi-Harada disease and Behcet's disease, and is essential for maintaining the dynamic homeostasis of autoimmunity, its exact role in uveitis remains unclear. In this study, diminished EGR2 expression was observed in both retinal microglia from experimental autoimmune uveitis (EAU) mice and inflammation-induced human microglia cell line (HMC3). We constructed a mice model with conditional knockout of EGR2 in microglia and found that EGR2 deficiency resulted in increased intraocular inflammation. Meanwhile, EGR2 overexpression downregulated the expression of inflammatory cytokines as well as cell migration and proliferation in HMC3 cells. Next, RNA sequencing and ChIP-PCR results indicated that EGR2 directly bound to its downstream target growth differentiation factor 15 (GDF15) and further regulated GDF15 transcription. Furthermore, intravitreal injection of GDF15 recombinant protein was shown to ameliorate EAU progression in vivo. Meanwhile, knockdown of GDF15 reversed the phenotype of EGR2 overexpression-induced microglial inflammation in vitro. In summary, this study highlighted the protective role of the transcription factor EGR2 in AU by modulating the microglial phenotype. GFD15 was identified as a downstream target of EGR2, providing a unique target for uveitis treatment.


Subject(s)
Autoimmune Diseases , Early Growth Response Protein 2 , Growth Differentiation Factor 15 , Microglia , Uveitis , Animals , Humans , Mice , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/metabolism , Cell Line , Disease Models, Animal , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/genetics , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Phenotype , Retina/metabolism , Retina/pathology , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology , Uveitis/genetics
2.
Adv Immunol ; 163: 21-49, 2024.
Article in English | MEDLINE | ID: mdl-39271258

ABSTRACT

As potent pro-inflammatory mediators, IL-17 family cytokines play crucial roles in the pathogenesis of various inflammatory and autoimmune skin disorders. Although substantial progress has been achieved in understanding the pivotal role of IL-17A signaling in psoriasis, leading to the development of highly effective biologics, the functions of other IL-17 family members in inflammatory or autoimmune skin diseases remain less explored. In this review, we provide a comprehensive overview of IL-17 family cytokines and their receptors, with a particular focus on the recent advancements in identifying cellular sources, receptors and signaling pathways regulated by these cytokines. At the end, we discuss how the aberrant functions of IL-17 family cytokines contribute to the pathogenesis of diverse inflammatory or autoimmune skin diseases.


Subject(s)
Autoimmune Diseases , Interleukin-17 , Signal Transduction , Skin Diseases , Humans , Interleukin-17/metabolism , Interleukin-17/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Animals , Skin Diseases/immunology , Skin Diseases/metabolism , Psoriasis/immunology , Psoriasis/metabolism , Inflammation/immunology , Inflammation/metabolism , Receptors, Interleukin-17/metabolism , Receptors, Interleukin-17/immunology
3.
Acta Biochim Biophys Sin (Shanghai) ; 56(8): 1145-1155, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39099413

ABSTRACT

Glycosylation, a crucial posttranslational modification, plays a significant role in numerous physiological and pathological processes. Lectin microarrays, which leverage the high specificity of lectins for sugar binding, are ideally suited for profiling the glycan spectra of diverse and complex biological samples. In this review, we explore the evolution of lectin detection technologies, as well as the applications and challenges of lectin microarrays in analyzing the glycome profiles of various clinical samples, including serum, saliva, tissues, sperm, and urine. This review not only emphasizes significant advancements in the high-throughput analysis of polysaccharides but also provides insight into the potential of lectin microarrays for diagnosing and managing diseases such as tumors, autoimmune diseases, and chronic inflammation. We aim to provide a clear, concise, and comprehensive overview of the use of lectin microarrays in clinical settings, thereby assisting researchers in conducting clinical studies in glycobiology.


Subject(s)
Glycomics , Lectins , Polysaccharides , Humans , Lectins/metabolism , Lectins/chemistry , Polysaccharides/metabolism , Polysaccharides/analysis , Glycomics/methods , Microarray Analysis/methods , Glycosylation , Neoplasms/metabolism , Neoplasms/diagnosis , Autoimmune Diseases/metabolism , Autoimmune Diseases/diagnosis
4.
Nat Cardiovasc Res ; 3(3): 301-316, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39196111

ABSTRACT

Myocarditis is an inflammatory heart disease that leads to loss of cardiomyocytes and frequently precipitates fibrotic remodeling of the myocardium, culminating in heart failure. However, the molecular mechanisms underlying immune cell control and maintenance of tissue integrity in the inflamed cardiac microenvironment remain elusive. In this study, we found that bone morphogenic protein-4 (BMP4) gradients maintain cardiac tissue homeostasis by single-cell transcriptomics analyses of inflamed murine and human myocardial tissues. Cardiac BMP pathway dysregulation was reflected by reduced BMP4 serum concentration in patients with myocarditis. Restoration of BMP signaling by antibody-mediated neutralization of the BMP inhibitors gremlin-1 and gremlin-2 ameliorated T cell-induced myocardial inflammation in mice. Moreover, progression to inflammatory cardiomyopathy was blocked through the reduction of fibrotic remodeling and preservation of cardiomyocyte integrity. These results unveil the BMP4-gremlin axis as a druggable pathway for the treatment of myocardial inflammation, limiting the severe sequelae of cardiac fibrosis and heart failure.


Subject(s)
Autoimmune Diseases , Bone Morphogenetic Protein 4 , Disease Models, Animal , Fibrosis , Myocarditis , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/immunology , Animals , Fibrosis/pathology , Fibrosis/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Humans , Autoimmune Diseases/pathology , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Male , Signal Transduction , Mice , Cellular Microenvironment , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Female , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Myocardium/immunology
5.
Sci Rep ; 14(1): 18829, 2024 08 13.
Article in English | MEDLINE | ID: mdl-39138267

ABSTRACT

Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a urinary disorder that affects youthful to middle-aged men most frequently. It has been revealed that Th17/Treg imbalance is a crucial factor in the pathophysiological mechanisms behind this disease. However, this imbalance's mechanisms are unknown. In the experimental autoimmune prostatitis (EAP) mouse model, the NLRP3 inflammasome was turned on, IL-1ß levels went up. Moreover, there exists a discernible positive association between the upsurge in IL-1ß and the perturbation of Th17/Treg equilibrium. Additionally, we have revealed that IL-1ß plays a vital role in promoting the differentiation of Naïve CD4+ T cells into the Th17 cells and enhances the conversion of Treg cells into Th17 cells. Further studies revealed that IL-1ß promotes STAT3 phosphorylation, which is what causes Treg cells to become Th17 cells. All data strongly suggest that the NLRP3 inflammatory influence Th17 cell development and the conversion of Treg cells into Th17 cells through IL-1ß, disrupting the Th17/Treg balance and exacerbating EAP inflammation. In this article, we provide new theories for the pathogenesis of CP/CPPS and propose new prevention and therapy methods.


Subject(s)
Autoimmune Diseases , Disease Models, Animal , Interleukin-1beta , NLR Family, Pyrin Domain-Containing 3 Protein , Prostatitis , T-Lymphocytes, Regulatory , Th17 Cells , Animals , Male , Prostatitis/immunology , Prostatitis/metabolism , Prostatitis/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-1beta/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Mice , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , STAT3 Transcription Factor/metabolism , Inflammasomes/metabolism , Cell Differentiation , Mice, Inbred C57BL
6.
Sci Rep ; 14(1): 19311, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164435

ABSTRACT

Autoimmune related kidney diseases (ARKDs), including minimal change nephropathy (MCN), membranous nephropathy (MN), IgA nephropathy (IgAN), and lupus nephritis (LN), significantly affect renal function. These diseases are characterized by the formation of local immune complexes and the subsequent activation of the complement system, leading to kidney damage and proteinuria. Despite the known patterns of glomerular injury, the specific molecular mechanisms that contribute to renal tubular damage across ARKDs remain underexplored. Laser capture microdissection and liquid chromatography-tandem mass spectrometry (LC-MS/MS) were used to conduct a comparative proteomic analysis of renal tubular tissues from formalin-fixed paraffin-embedded samples. The cohort comprised of 10 normal controls (NC), 5 MCN, 4 MN, 17 IgAN, and 21 LN patients. Clinical parameters and histopathological assessments were integrated with proteomic findings to comprehensively investigate underlying pathogenic processes. Clinical evaluation indicated significant glomerular damage, as reflected by elevated urinary protein levels and reduced plasma albumin levels in patients with ARKD. Histological analyses confirmed varying degrees of tubular damage and deposition of immune complexes. Proteomic analyses identified significant changes in protein expression, particularly in complement components (C3, C4A, C4B, C8G, CFB, and SERPINA1) and mitochondrial proteins (ATP5F1E and ATP5PD), highlighting the common alterations in the complement system and mitochondrial proteins across ARKDs. These alterations suggest a novel complement-mitochondrial-epithelial-mesenchymal transition (EMT) pathway axis that contributes to tubular damage in ARKDs. Notably, significant alterations in CFB in tubular ARKD patients were revealed, implicating it as a therapeutic target. This study underscores the importance of complement activation and mitochondrial dysfunction in the pathogenesis of ARKDs, and proposes CFB as a potential therapeutic target to inhibit complement activation and mitigate tubular damage. Future research should validate the complement-mitochondrial-EMT pathway axis and explore the effects and mechanisms of CFB inhibitors in alleviating ARKD progression.


Subject(s)
Complement Activation , Mitochondria , Proteomics , Humans , Proteomics/methods , Female , Male , Adult , Mitochondria/metabolism , Middle Aged , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Tandem Mass Spectrometry , Laser Capture Microdissection , Complement System Proteins/metabolism , Chromatography, Liquid
7.
Life Sci ; 355: 122996, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39173995

ABSTRACT

AIMS: To investigate the therapeutic potential of visual stimulation (VS) and BDNF in murine experimental autoimmune uveoretinitis (EAU). MAIN METHODS: Mice were immunized by subcutaneous injection of interphotoreceptor retinoid-binding protein in Freund's complete adjuvant and intravenous injection of pertussis toxin, and were then exposed to high-contrast VS 12 h/day (days 1-14 post-immunization). EAU severity was assessed by examining clinical score, visual acuity, inflammatory markers, and immune cells in the retina. The transcriptome of activated retinal cells was determined by RNA-seq using RNA immunoprecipitated in complex with phosphorylated ribosomal protein S6. The retinal levels of protein products of relevant upregulated genes were quantified. The effect of BDNF on EAU was tested in unstimulated mice by its daily topical ocular administration (days 8-14 post-immunization). KEY FINDINGS: VS attenuated EAU development and decreased the expression of pro-inflammatory cytokines/chemokines and numbers of immune cells in the retina (n = 10-20 eyes/group for each analysis). In activated retinal cells of control mice (n = 30 eyes/group), VS upregulated genes encoding immunomodulatory neuropeptides, of which BDNF and vasoactive intestinal peptide (VIP) also showed increased mRNA and protein levels in the retina of VS-treated EAU mice (n = 6-10 eyes/group for each analysis). In unstimulated EAU mice, BDNF treatment mimicked the protective effects of VS by modulating the inflammatory and stem cell properties of Müller cells (n = 5 eyes/group for each analysis). SIGNIFICANCE: VS effectively suppresses EAU, at least through enhancing retinal levels of anti-inflammatory and neuroprotective factors, VIP and BDNF. Our findings also suggest BDNF as a promising therapeutic agent for uveitis treatment.


Subject(s)
Autoimmune Diseases , Brain-Derived Neurotrophic Factor , Retinitis , Uveitis , Animals , Mice , Brain-Derived Neurotrophic Factor/metabolism , Uveitis/metabolism , Uveitis/drug therapy , Uveitis/immunology , Retinitis/drug therapy , Retinitis/prevention & control , Retinitis/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Female , Retina/metabolism , Retina/drug effects , Mice, Inbred C57BL , Vasoactive Intestinal Peptide/pharmacology , Disease Models, Animal , Cytokines/metabolism
8.
J Mass Spectrom ; 59(9): e5083, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39162140

ABSTRACT

Protein glycosylation is the co- and/or post-translational modification of proteins with oligosaccharides (glycans). This process is not template based and can introduce a heterogeneous set of glycan modifications onto substrate proteins. Glycan structures preserve biomolecular information from the cell, with glycoproteins from different cell types and tissues displaying distinct patterns of glycosylation. Several decades of research have revealed that glycan structures also differ between normal physiology and disease. This suggests that the information stored in glycoproteins and glycans can be utilized for disease diagnosis and monitoring. Methods that enable sensitive and site-specific measurement of protein glycosylation in clinical settings, such as nano-flow liquid chromatography tandem mass spectrometry, are therefore essential. The purpose of this perspective is to discuss recent advances in mass spectrometry and the potential of these advances to facilitate the detection and monitoring of disease-specific glycoprotein glycoforms. Glycoproteomics, the system-wide characterization of glycoprotein identity inclusive of site-specific characterization of carbohydrate modifications on proteins, and glycomics, the characterization of glycan structures, will be discussed in this context. Quantitative measurement of glycopeptide markers via parallel reaction monitoring is highlighted. The development of promising glycopeptide markers for autoimmune disease, liver disease, and liver cancer is discussed. Synthetic glycopeptide standards, ambient ionization mass spectrometry, and consideration of glyco-biomarkers in two- and three-dimensional space within tissue will be critical to the advancement of this field. The authors envision a future in which glycoprotein mass spectrometry workflows will be integrated into clinical settings, to aid in the rapid diagnosis and monitoring of disease.


Subject(s)
Glycoproteins , Polysaccharides , Proteomics , Humans , Glycoproteins/analysis , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation , Proteomics/methods , Polysaccharides/analysis , Polysaccharides/chemistry , Biomarkers/analysis , Mass Spectrometry/methods , Glycomics/methods , Glycopeptides/analysis , Glycopeptides/chemistry , Protein Processing, Post-Translational , Tandem Mass Spectrometry/methods , Autoimmune Diseases/diagnosis , Autoimmune Diseases/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/chemistry , Liver Diseases/diagnosis , Liver Diseases/metabolism , Chromatography, Liquid/methods
9.
Front Biosci (Landmark Ed) ; 29(8): 301, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39206903

ABSTRACT

The chemokine receptor CXCR3 and its ligands (MIG/CXCL9, IP-10/CXCL10, and I-TAC/CXCL11) play a central role in the generation of cellular inflammation, both in the protective responses to invading pathogens, and in different pathological conditions associated with autoimmunity. It is worth noting that CXCR3 is highly expressed on innate and adaptive lymphocytes, as well as on various cell subsets that are localized in non-immune organs and tissues. Our review focuses exclusively on CXCR3-expressing T cells, including Th1, Th17.1, Tfh17, Tfh17.1, CXCR3+ Treg cells, and Tc1 CD8+ T cells. Currently, numerous studies have highlighted the role of CXCR3-dependent interactions in the coordination of inflammation in the peripheral tissues, both to increase recruitment of CD4+ and CD8+ T cells that upregulate inflammation, and also for recruitment of CXCR3+ T regulatory cells to dampen overexuberant responses. Understanding the role of CXCR3 and its ligands might help to apply them as new and effective therapeutic targets in a wide range of diseases.


Subject(s)
Autoimmunity , Receptors, CXCR3 , Receptors, CXCR3/metabolism , Receptors, CXCR3/immunology , Humans , Autoimmunity/immunology , Animals , Infections/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Inflammation/immunology , Inflammation/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism
10.
Eur J Med Chem ; 277: 116783, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39180944

ABSTRACT

Cytokines play a crucial role in the pathophysiology of autoimmune and inflammatory diseases, with over 50 cytokines undergoing signal transduction through the Signal Transducers and Activators of Transcription (STAT) signaling pathway. Recent studies have solidly confirmed the pivotal role of STATs in autoimmune and inflammatory diseases. Therefore, this review provides a detailed summary of the immunological functions of STATs, focusing on exploring their mechanisms in various autoimmune and inflammatory diseases. Additionally, with the rapid advancement of structural biology in the field of drug discovery, many STAT inhibitors have been identified using structure-based drug design strategies. In this review, we also examine the structures of STAT proteins and compile the latest research on STAT inhibitors currently being tested in animal models and clinical trials for the treatment of immunological diseases, which emphasizes the feasibility of STATs as promising therapeutic targets and provides insights into the design of the next generation of STAT inhibitors.


Subject(s)
Autoimmune Diseases , Inflammation , STAT Transcription Factors , Humans , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , Animals , Inflammation/drug therapy , Inflammation/metabolism , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Molecular Structure
11.
Sci Rep ; 14(1): 20033, 2024 08 28.
Article in English | MEDLINE | ID: mdl-39198470

ABSTRACT

Uveitis is characterised by breakdown of the blood-retinal barrier (BRB), allowing infiltration of immune cells that mediate intraocular inflammation, which can lead to irreversible damage of the neuroretina and the loss of sight. Treatment of uveitis relies heavily on corticosteroids and systemic immunosuppression due to limited understanding of disease pathogenesis. We performed single-cell RNA-sequencing of retinas, as well as bulk RNA-sequencing of retinal pigment epithelial (RPE) cells from mice with experimental autoimmune uveitis (EAU) versus healthy control. This revealed that the Th1/Th17-driven disease induced strong gene expression changes in response to inflammation in rods, cones, Müller glia and RPE. In particular, Müller glia and RPE cells were found to upregulate expression of chemokines, complement factors, leukocyte adhesion molecules and MHC class II, thus highlighting their contributions to immune cell recruitment and antigen presentation at the inner and outer BRB, respectively. Additionally, ligand-receptor interaction analysis with CellPhoneDB revealed key interactions between Müller glia and T cell / natural killer cell subsets via chemokines, galectin-9 to P4HB/TIM-3, PD-L1 to PD-1, and nectin-2/3 to TIGIT signalling axes. Our findings elucidate mechanisms contributing to breakdown of retinal immune privilege during uveitis and identify novel targets for therapeutic interventions.


Subject(s)
Autoimmune Diseases , Blood-Retinal Barrier , Single-Cell Analysis , Uveitis , Animals , Uveitis/immunology , Uveitis/genetics , Uveitis/metabolism , Uveitis/pathology , Blood-Retinal Barrier/metabolism , Mice , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Disease Models, Animal , Retina/metabolism , Retina/immunology , Retina/pathology , Retinal Pigment Epithelium/metabolism , Transcriptome , Gene Expression Profiling , Ependymoglial Cells/metabolism , Mice, Inbred C57BL
12.
Biomolecules ; 14(8)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39199429

ABSTRACT

N6-methyladenosine (m6A) represents the most prevalent and significant internal modification in mRNA, with its critical role in gene expression regulation and cell fate determination increasingly recognized in recent research. The immune system, essential for defense against infections and maintaining internal stability through interactions with other bodily systems, is significantly influenced by m6A modification. This modification acts as a key post-transcriptional regulator of immune responses, though its effects on different immune cells vary across diseases. This review delineates the impact of m6A modification across major system-related cancers-including those of the respiratory, digestive, endocrine, nervous, urinary reproductive, musculoskeletal system malignancies, as well as acute myeloid leukemia and autoimmune diseases. We explore the pathogenic roles of m6A RNA modifications within the tumor immune microenvironment and the broader immune system, highlighting how RNA modification regulators interact with immune pathways during disease progression. Furthermore, we discuss how the expression patterns of these regulators can influence disease susceptibility to immunotherapy, facilitating the development of diagnostic and prognostic models and pioneering new therapeutic approaches. Overall, this review emphasizes the challenges and prospective directions of m6A-related immune regulation in various systemic diseases throughout the body.


Subject(s)
Adenosine , Neoplasms , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/metabolism , Methylation , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/metabolism
13.
Brain Behav Immun ; 122: 241-255, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39084540

ABSTRACT

Pediatric Acute-onset Neuropsychiatric Syndrome (PANS) is characterized by the abrupt onset of significant obsessive-compulsive symptoms (OCS) and/or severe food restriction, together with other neuropsychiatric manifestations. An autoimmune pathogenesis triggered by infection has been proposed for at least a subset of PANS. The older diagnosis of Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcus (PANDAS) describes rapid onset of OCD and/or tics associated with infection with Group A Streptococcus. The pathophysiology of PANS and PANDAS remains incompletely understood. We recently found serum antibodies from children with rigorously defined PANDAS to selectively bind to cholinergic interneurons (CINs) in the striatum. Here we examine this binding in children with relapsing and remitting PANS, a more heterogeneous condition, collected in a distinct clinical context from those examined in our previous work, from children with a clinical history of Streptococcus infection. IgG from PANS cases showed elevated binding to striatal CINs in both mouse and human brain. Patient plasma collected during symptom flare decreased a molecular marker of CIN activity, phospho-riboprotein S6, in ex vivo brain slices; control plasma did not. Neither elevated antibody binding to CINs nor diminished CIN activity was seen with plasma collected from the same children during remission. These findings replicate what we have seen previously in PANDAS and support the hypothesis that at least a subset of PANS cases have a neuroimmune pathogenesis. Given the critical role of CINs in modulating basal ganglia function, these findings confirm striatal CINs as a locus of interest in the pathophysiology of both PANS and PANDAS.


Subject(s)
Corpus Striatum , Interneurons , Obsessive-Compulsive Disorder , Streptococcal Infections , Humans , Child , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Male , Obsessive-Compulsive Disorder/metabolism , Obsessive-Compulsive Disorder/immunology , Female , Animals , Interneurons/metabolism , Interneurons/immunology , Mice , Corpus Striatum/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Adolescent , Immunoglobulin G/metabolism , Autoantibodies/metabolism , Autoantibodies/immunology , Cholinergic Neurons/metabolism , Child, Preschool
14.
Int J Biol Macromol ; 278(Pt 1): 134193, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39069042

ABSTRACT

Long non-coding RNAs (lncRNAs) have been implicated in dilated cardiomyopathy (DCM). However, the biological functions and regulatory mechanisms of lncRNAs in DCM remain elusive. Using a mouse model of experimental autoimmune myocarditis (EAM) to mimic DCM, we successfully constructed a dynamic lncRNA expression library for EAM by lncRNA microarray and found that the expression of a macrophage-enriched lncRNA, MAAMT, was significantly increased in the myocardial tissue of mice at the acute stage of EAM. Functionally, MAAMT knockdown alleviated the recruitment and proinflammatory activation of macrophages in the heart, spleen, and peripheral blood of mice at the acute stage of EAM, reduced myocardial inflammation and injury, and eventually reversed ventricular remodelling and improved cardiac function in mice at the chronic stage of EAM. Mechanistically, we identified serine/arginine-rich splicing factor 1 (SRSF1) as an MAAMT-interacting protein in macrophages using RNA pull-down assays coupled with mass spectrometry. MAAMT knockdown attenuated the ubiquitination-mediated degradation of SRSF1, increased the protein expression of SRSF1, and restrained the activation of the NF-κB pathway in macrophages, thereby inhibiting the proinflammatory activation of macrophages. Collectively, our results demonstrate that MAAMT is a key proinflammatory regulator of myocarditis that promotes macrophage activation through the SRSF1-NF-κB axis, providing a new insight into early effective treatment strategies for DCM.


Subject(s)
Autoimmune Diseases , Macrophages , Myocarditis , NF-kappa B , RNA, Long Noncoding , Serine-Arginine Splicing Factors , Signal Transduction , Animals , Myocarditis/metabolism , Myocarditis/immunology , Myocarditis/genetics , Myocarditis/pathology , RNA, Long Noncoding/genetics , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Mice , NF-kappa B/metabolism , Macrophages/metabolism , Macrophages/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Male , Disease Models, Animal , Macrophage Activation , Inflammation/genetics , Inflammation/metabolism
15.
Front Immunol ; 15: 1390907, 2024.
Article in English | MEDLINE | ID: mdl-38962006

ABSTRACT

Autoimmune diseases (AID) have emerged as prominent contributors to disability and mortality worldwide, characterized by intricate pathogenic mechanisms involving genetic, environmental, and autoimmune factors. In response to this challenge, a growing body of research in recent years has delved into genetic modifications, yielding valuable insights into AID prevention and treatment. Sirtuins (SIRTs) constitute a class of NAD-dependent histone deacetylases that orchestrate deacetylation processes, wielding significant regulatory influence over cellular metabolism, oxidative stress, immune response, apoptosis, and aging through epigenetic modifications. Resveratrol, the pioneering activator of the SIRTs family, and its derivatives have captured global scholarly interest. In the context of AID, these compounds hold promise for therapeutic intervention by modulating the SIRTs pathway, impacting immune cell functionality, suppressing the release of inflammatory mediators, and mitigating tissue damage. This review endeavors to explore the potential of resveratrol and its derivatives in AID treatment, elucidating their mechanisms of action and providing a comprehensive analysis of current research advancements and obstacles. Through a thorough examination of existing literature, our objective is to advocate for the utilization of resveratrol and its derivatives in AID treatment while offering crucial insights for the formulation of innovative therapeutic approaches.


Subject(s)
Autoimmune Diseases , Resveratrol , Sirtuins , Resveratrol/therapeutic use , Resveratrol/pharmacology , Humans , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Animals , Sirtuins/metabolism
16.
Cell Death Dis ; 15(7): 481, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965216

ABSTRACT

Autoimmune diseases commonly affect various systems, but their etiology and pathogenesis remain unclear. Currently, increasing research has highlighted the role of ferroptosis in immune regulation, with immune cells being a crucial component of the body's immune system. This review provides an overview and discusses the relationship between ferroptosis, programmed cell death in immune cells, and autoimmune diseases. Additionally, it summarizes the role of various key targets of ferroptosis, such as GPX4 and TFR, in immune cell immune responses. Furthermore, the release of multiple molecules, including damage-associated molecular patterns (DAMPs), following cell death by ferroptosis, is examined, as these molecules further influence the differentiation and function of immune cells, thereby affecting the occurrence and progression of autoimmune diseases. Moreover, immune cells secrete immune factors or their metabolites, which also impact the occurrence of ferroptosis in target organs and tissues involved in autoimmune diseases. Iron chelators, chloroquine and its derivatives, antioxidants, chloroquine derivatives, and calreticulin have been demonstrated to be effective in animal studies for certain autoimmune diseases, exerting anti-inflammatory and immunomodulatory effects. Finally, a brief summary and future perspectives on the research of autoimmune diseases are provided, aiming to guide disease treatment strategies.


Subject(s)
Autoimmune Diseases , Ferroptosis , Iron , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Iron/metabolism , Animals , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology
17.
Adv Protein Chem Struct Biol ; 142: 421-436, 2024.
Article in English | MEDLINE | ID: mdl-39059993

ABSTRACT

Host-pathogen interactions are complex associations which evolve over long co-evolutionary histories. Pathogens exhibit different mechanisms to gain advantage over their host. Mimicry of host factors is an influential tool in subverting host mechanisms to ensure pathogenesis. This chapter discusses such molecular mimicry exhibited during viral infections. Understanding the evolutionary relationships, shared identity and functional impact of the virus encoded mimics is critical. With a particular emphasis on viral mimics and their association with cancer and autoimmune diseases, this chapter highlights the importance of molecular mimicry in virus biology.


Subject(s)
Molecular Mimicry , Humans , Viruses/metabolism , Host-Pathogen Interactions , Virus Diseases/metabolism , Virus Diseases/virology , Virus Diseases/immunology , Endocrine System/metabolism , Neoplasms/metabolism , Neoplasms/virology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/virology , Autoimmune Diseases/immunology
18.
Int J Biol Sci ; 20(9): 3393-3411, 2024.
Article in English | MEDLINE | ID: mdl-38993566

ABSTRACT

Chronic prostatitis is one of the most common urologic diseases that troubles young men, with unclear etiology and ineffective treatment approach. Pyroptosis is a novel model of cell death, and its roles in chronic prostatitis are unknown. In this study, P2X7R, NEK7, and GSDMD-NT expression levels were detected in prostate tissues from benign prostate hyperplasia (BPH) patients and experiment autoimmune prostatitis (EAP) mice. P2X7R agonist, antagonist, NLRP3 inhibitor, and disulfiram were used to explore the roles of the P2X7R-NEK7-NLRP3 axis in prostate epithelial cell pyroptosis and chronic prostatitis development. We found that P2X7R, NEK7, and GSDMD-NT were highly expressed in the prostate epithelial cells of BPH patients with prostatic inflammation and EAP mice. Activation of P2X7R exacerbated prostatic inflammation and increased NLRP3 inflammasome component expressions and T helper 17 (Th17) cell proportion. Moreover, P2X7R-mediated potassium efflux promoted NEK7-NLRP3 interaction, and NLRP3 assembly and activation, which caused GSDMD-NT-mediated prostate epithelial cell pyroptosis to exacerbate EAP development. Disulfiram could effectively improve EAP by inhibiting GSDMD-NT-mediated prostate epithelial cell pyroptosis. In conclusion, the P2X7R-NEK7-NLRP3 axis could promote GSDMD-NT-mediated prostate epithelial cell pyroptosis and chronic prostatitis development, and disulfiram may be an effective drug to treat chronic prostatitis.


Subject(s)
Epithelial Cells , NIMA-Related Kinases , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphate-Binding Proteins , Prostate , Prostatitis , Pyroptosis , Animals , Humans , Male , Mice , Autoimmune Diseases/metabolism , Epithelial Cells/metabolism , Gasdermins , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Prostate/metabolism , Prostatitis/metabolism , Pyroptosis/drug effects , Receptors, Purinergic P2X7/metabolism
19.
Exp Dermatol ; 33(7): e15135, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39021278

ABSTRACT

Autoimmune skin disease is a kind of heterogeneous disease with complicated pathogenesis. Many factors such as genetic, infectious, environmental and even psychological factors may interact together to trigger a synergistic effect for the development of abnormal innate and adaptive immune responses. Although the exact mechanisms remain unclear, recent evidence suggests that pyroptosis plays a pivotal role in the development of autoimmune skin disease. The feature of pyroptosis is the first formation of pores in cellular membranes, then cell rupture and the release of intracellular substances and pro-inflammatory cytokines, such as interleukin-1 beta (IL-1ß) and IL-18. This hyperactive inflammatory programmed cell death damages the homeostasis of the immune system and advances autoimmunity. This review briefly summarises the molecular regulatory mechanisms of pyrin domain-containing protein 3 (NLRP3) inflammasome and gasdermin family, as well as the molecular mechanisms of pyroptosis, highlights the latest progress of pyroptosis in autoimmune skin disease, including systemic lupus erythematosus, psoriasis, atopic dermatitis and systemic scleroderma and attempts to identify its potential advantages as a therapeutic target or prognostic biomarker for these diseases.


Subject(s)
Autoimmune Diseases , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Skin Diseases/immunology , Animals , Phosphate-Binding Proteins/metabolism , Interleukin-1beta/metabolism , Scleroderma, Systemic/immunology , Lupus Erythematosus, Systemic/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Psoriasis/immunology , Psoriasis/metabolism , Autoimmunity , Interleukin-18/metabolism , Dermatitis, Atopic/immunology
20.
Molecules ; 29(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38998954

ABSTRACT

In recent years, phage display technology has become vital in clinical research. It helps create antibodies that can specifically bind to complex antigens, which is crucial for identifying biomarkers and improving diagnostics and treatments. However, existing reviews often overlook its importance in areas outside cancer research. This review aims to fill that gap by explaining the basics of phage display and its applications in detecting and treating various non-cancerous diseases. We focus especially on its role in degenerative diseases, inflammatory and autoimmune diseases, and chronic non-communicable diseases, showing how it is changing the way we diagnose and treat illnesses. By highlighting important discoveries and future possibilities, we hope to emphasize the significance of phage display in modern healthcare.


Subject(s)
Biomarkers , Cell Surface Display Techniques , Humans , Noncommunicable Diseases/epidemiology , Peptide Library , Autoimmune Diseases/diagnosis , Autoimmune Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL