Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.499
1.
Cytokine ; 169: 156268, 2023 09.
Article En | MEDLINE | ID: mdl-37320965

Cytokines are a protein family comprising interleukins, lymphokines, chemokines, monokines and interferons. They are significant constituents of the immune system, and they act in accordance with specific cytokine inhibiting compounds and receptors for the regulation of immune responses. Cytokine studies have resulted in the establishment of newer therapies which are being utilized for the treatment of several malignant diseases. The advancement of these therapies has occurred from two distinct strategies. The first strategy involves administrating the recombinant and purified cytokines, and the second strategy involves administrating the therapeutics which inhibits harmful effects of endogenous and overexpressed cytokines. Colony stimulating factors and interferons are two exemplary therapeutics of cytokines. An important effect of cytokine receptor antagonist is that they can serve as anti-inflammatory agents by altering the treatments of inflammation disorder, therefore inhibiting the effects of tumour necrosis factor. In this article, we have highlighted the research behind the establishment of cytokines as therapeutics and vaccine adjuvants, their role of immunotolerance, and their limitations.


Adjuvants, Vaccine , Cytokines , Cytokines/metabolism , Interferons/physiology , Immunologic Factors , Interleukins/physiology , Immunotherapy
2.
PLoS One ; 17(12): e0279584, 2022.
Article En | MEDLINE | ID: mdl-36548354

Mesenchymal stem cells (MSC) can differentiate into chondrocytes. Epstein-Barr virus-induced gene 3 (EBI3) is differentially expressed during chondrogenic differentiation and can be produced by MSC. EBI3 is also a subunit of interleukin (IL)-27 and IL-35, and it accumulates in the endoplasmic reticulum (ER) when its partners, such as IL-27 p28 and IL-35 p35, are insufficient. ER stress induced by protein accumulation is responsible for chondrogenic differentiation. However, the role of EBI3 and its relevance to the ER stress in chondrogenic differentiation of MSC have never been addressed. Here, we demonstrate that EBI3 protein is expressed in the early stage of chondrogenic differentiation of MSC. Additionally, knockdown, overexpression, or induction of EBI3 through IL-1ß inhibits chondrogenesis. We show that EBI3 localizes and accumulates in the ER of MSC after overexpression or induction by IL-1ß and TNF-α, whereas ER stress inhibitor 4-phenylbutyric acid decreases its accumulation in MSC. Moreover, EBI3 modulates ER stress sensor inositol-requiring enzyme 1 α (IRE1α) after induced by IL-1ß, and MSC-like cells coexpress EBI3 and IRE1α in rheumatoid arthritis (RA) synovial tissue. Altogether, these data demonstrate that intracellular EBI3 commits to chondrogenic differentiation by regulating ER stress sensor IRE1α.


Cell Differentiation , Chondrocytes , Chondrogenesis , Endoplasmic Reticulum Stress , Interleukins , Mesenchymal Stem Cells , Minor Histocompatibility Antigens , Humans , Chondrocytes/cytology , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Interleukins/genetics , Interleukins/physiology , Mesenchymal Stem Cells/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/physiology , Cell Differentiation/genetics , Chondrogenesis/genetics
3.
Dermatology ; 238(5): 829-836, 2022.
Article En | MEDLINE | ID: mdl-35537419

Twenty years after the cloning, characterization, and identification of interleukin (IL)-22 in 2000, the precise biological role of this cytokine in healthy and unhealthy skin is not completely known. The aim of this review is to provide an overview on the recent knowledge available in literature about the origin, sources, targets, molecular mechanism of action, and clinical issues regarding IL-22. Last but not least, recent experimental evidence obtained in a 3D model of organotypic culture of normal human skin highlights its homeostatic role and will be discussed in detail, as personal observations. As most of the data concerning IL-22 immunomodulating activity are obtained from mouse models, this work offers a new perspective on its clinical role. The hypothesis herein advanced is that IL-22 profoundly affects keratinocyte terminal differentiation, whereas, in order to induce a proliferation impairment, a more complex psoriatic-like microenvironment is needed.


Epidermis/physiology , Interleukins/physiology , Skin Diseases , Animals , Homeostasis , Humans , Keratinocytes , Mice , Psoriasis , Skin , Skin Diseases/physiopathology , Interleukin-22
4.
Oncoimmunology ; 11(1): 2057837, 2022.
Article En | MEDLINE | ID: mdl-35371618

Multiple myeloma (MM) is characterized by an accumulation of monoclonal plasma cells within the bone marrow (BM). Macrophages are an abundant component of myeloma BM microenvironment and support survival of the malignant cells and foster myeloma development and progression by suppression of the immune response. In our previous study, we found that MM patients overexpress pro-inflammatory cytokine interleukin-32 (IL-32). The present study aimed to investigate the role of IL-32 in myeloma progression and mechanisms of IL-32 on macrophages functions. We discovered that the expression of IL-32 was associated with the disease stage in myeloma patients. MM-derived exosomes containing IL-32γ promoted the expression of programmed death-ligand 1(PD-L1) by macrophages, thus promoting immune evasion. Mechanistically, myeloma-secreted IL-32γ acted via proteinase 3 (PR3) to enhance 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) dependent glycolysis and subsequent PD-L1 expression. Moreover, the PFKFB3-Janus kinase 1 (JAK1) axis might contribute to the expression of PD-L1 by macrophages. To sum up, we concluded that IL-32 was a critical mediator in myeloma progression, and targeting IL-32 signaling might be used as a potential strategy for treating myeloma.


B7-H1 Antigen , Interleukins , Multiple Myeloma , B7-H1 Antigen/genetics , Humans , Interleukins/physiology , Janus Kinase 1/metabolism , Macrophages/metabolism , Multiple Myeloma/metabolism , Phosphofructokinase-2/metabolism , Tumor Microenvironment
5.
Ital J Dermatol Venerol ; 157(4): 306-312, 2022 Aug.
Article En | MEDLINE | ID: mdl-35274883

Chronic inflammatory skin diseases pose significant challenges for both patients and clinicians worldwide. Atopic dermatitis (AD), the most common of these diseases, affects up to 8% of the adult population depending on geographic location and demographic group, while prurigo nodularis (PN) is a less common disease that causes significant burden. In these inflammatory skin conditions, pruritus is a cardinal symptom. Interleukin 31 (IL-31), described as a neuroimmune modulator, has been shown to have a prominent role in both inflammation and itch. IL-31 acts through a receptor complex consisting of IL-31 receptor α (IL-31RA) and oncostatin M receptor ß (OSMRß). IL-31 is produced by a variety of cells, including type 2 helper T cells, and IL-31 signaling can activate three important pathways: JAK/STAT, P13K/AKT, and ERK/MAPK. IL-31 is elevated in AD and PN, and is thought to induce chemokine genes CCL1, CCL17, and CCL22. The chemokines recruit T cells to affected skin, where more IL-31 is secreted. The IL-31 receptor complex is also abundant in dorsal root ganglia in human tissue, home of primary sensory neurons and the distal source of "itch sensations." IL-31 and its receptor complex have an important role in chronic inflammatory diseases, including AD and PN, and blocking the IL-31/IL-31RA signaling may represent an important new therapeutic approach for these diseases, which continue to have significant unmet medical needs.


Cytokines , Dermatitis, Atopic , Interleukins , Cytokines/physiology , Dermatitis, Atopic/drug therapy , Humans , Interleukins/physiology , Pruritus , Receptors, Interleukin/physiology , Skin
6.
Front Immunol ; 13: 833636, 2022.
Article En | MEDLINE | ID: mdl-35185925

The establishment of an "interferon (IFN) signature" to subset SLE patients on disease severity has led to therapeutics targeting IFNα. Here, we investigate IFN signaling in SLE using multiplexed protein arrays and single cell cytometry by time of flight (CyTOF). First, the IFN signature for SLE patients (n=81) from the Stanford Lupus Registry is determined using fluidigm qPCR measuring 44 previously determined IFN-inducible transcripts. IFN-high (IFN-H) patients have increased SLE criteria and renal/CNS/immunologic involvement, and increased autoantibody reactivity against spliceosome-associated antigens. CyTOF analysis is performed on non-stimulated and stimulated (IFNα, IFNγ, IL-21) PBMCs from SLE patients (n=25) and HCs (n=9) in a panel identifying changes in phosphorylation of intracellular signaling proteins (pTOF). Another panel is utilized to detect changes in intracellular cytokine (ICTOF) production in non-stimulated and stimulated (PMA/ionomycin) PBMCs from SLE patients (n=31) and HCs (n=17). Bioinformatic analysis by MetaCyto and OMIQ reveal phenotypic changes in immune cell subsets between IFN-H and IFN-low (IFN-L) patients. Most notably, IFN-H patients exhibit increased STAT1/3/5 phosphorylation downstream of cytokine stimulation and increased phosphorylation of non-canonical STAT proteins. These results suggest that IFN signaling in SLE modulates STAT phosphorylation, potentially uncovering possible targets for future therapeutic approaches.


Interferon Type I/physiology , Interleukins/physiology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/metabolism , STAT1 Transcription Factor/metabolism , Adult , Female , Flow Cytometry , Humans , Interferon Type I/analysis , Interleukins/analysis , Male , Middle Aged , Phosphorylation , Signal Transduction , Single-Cell Analysis
7.
Int J Mol Sci ; 23(4)2022 Feb 16.
Article En | MEDLINE | ID: mdl-35216313

Recurrent implantation failure (RIF) is a multifactorial condition affecting 10-15% of in vitro fertilization (IVF) couples. Data suggest that functional dysregulation of the endometrial immune system constitutes one of the main pathophysiological mechanisms leading to RIF. The aim of this article is to provide a thorough presentation and evaluation of the role of interleukins (ILs) in the pathogenesis of RIF. A comprehensive literature screening was performed summarizing current evidence. During implantation, several classes of ILs are secreted by epithelial and stromal endometrial cells, including IL-6, IL-10, IL-12, IL-15, IL-18, and the leukemia inhibitory factor. These ILs create a perplexing network that orchestrates both proliferation and maturation of uterine natural killer cells, controls the function of regulatory T and B cells inhibiting the secretion of antifetal antibodies, and supports trophoblast invasion and decidua formation. The existing data indicate associations between ILs and RIF. The extensive analysis performed herein concludes that the dysregulation of the ILs network indeed jeopardizes implantation leading to RIF. This review further proposes a mapping of future research on how to move forward from mere associations to robust molecular data that will allow an accurate profiling of ILs in turn enabling evidence-based consultancy and decision making when addressing RIF patients.


Embryo Implantation , Endometrium , Interleukins , Embryo Implantation/physiology , Endometrium/pathology , Female , Fertilization in Vitro , Humans , Infertility, Female , Interleukins/physiology , Uterus
8.
Front Immunol ; 12: 766586, 2021.
Article En | MEDLINE | ID: mdl-34868019

Cytokines are powerful mediators of inflammation. Consequently, their potency is regulated in many ways to protect the host. Several cytokines, including IL-22, have coordinating binding proteins or soluble receptors that bind to the cytokine, block the interaction with the cellular receptor, and thus prevent cellular signaling. IL-22 is a critical cytokine in the modulation of tissue responses during inflammation and is highly upregulated in many chronic inflammatory disease patients, including those with psoriasis, rheumatoid arthritis, and inflammatory bowel disease (IBD). In healthy individuals, low levels of IL-22 are secreted by immune cells, mainly in the gastrointestinal (GI) tract. However, much of this IL-22 is likely not biologically active due to the high levels of IL-22 binding protein (IL-22BP) produced by intestinal dendritic cells (DCs). IL-22BP is a soluble receptor homolog that binds to IL-22 with greater affinity than the membrane spanning receptor. Much is known regarding the regulation and function of IL-22 in health and disease. However, less is known about IL-22BP. In this review, we will focus on IL-22BP, including its regulation, role in IL-22 biology and inflammation, and promise as a therapeutic. IL-22 can be protective or pathogenic, depending on the context of inflammation. IL-22BP also has divergent roles. Ongoing and forthcoming studies will expand our knowledge of IL-22BP and IL-22 biology, and suggest that IL-22BP holds promise as a way to regulate IL-22 biology in patients with chronic inflammatory disease.


Interleukins/physiology , Receptors, Interleukin/physiology , Gastrointestinal Tract/immunology , Humans , Inflammation/immunology , Liver/immunology , Receptors, Interleukin/analysis , Skin/immunology , Interleukin-22
9.
J Interferon Cytokine Res ; 41(11): 391-406, 2021 11.
Article En | MEDLINE | ID: mdl-34788131

The balance between inflammatory and anti-inflammatory immune responses is maintained through immunoregulatory cell populations and immunosuppressive cytokines. Interleukin-35 (IL-35), an inhibitory cytokine that belongs to the IL-12 family, is capable of potently suppressing T cell proliferation and inducing IL-35-producing induced regulatory T cells (iTr35) to limit inflammatory responses. Over the past decade, a growing number of studies have indicated that IL-35 plays an important role in controlling immune-related disorders, including autoimmune diseases, infectious diseases, and cancer. In this review, we summarize the current knowledge about the biology of IL-35 and its contribution in different diseases, and we discuss the potential of and barriers to harnessing IL-35 as a clinical biomarker or immunotherapy.


Inflammation/metabolism , Interleukins/chemistry , Interleukins/physiology , Animals , Arthritis, Rheumatoid/metabolism , Atherosclerosis/metabolism , Autoimmune Diseases/immunology , Autoimmunity , Biomarkers/metabolism , Cell Proliferation , Communicable Diseases/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Expression Regulation , Humans , Immune System Diseases , Immunotherapy , Inflammatory Bowel Diseases/metabolism , Interleukin-12 Subunit p35/blood , Interleukins/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation , Multiple Sclerosis/metabolism , Neoplasms/metabolism , Signal Transduction , T-Lymphocytes/chemistry , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
10.
Front Immunol ; 12: 754413, 2021.
Article En | MEDLINE | ID: mdl-34737752

Eosinophils are leukocytes which reside in the gastrointestinal tract under homeostatic conditions, except for the esophagus which is normally devoid of eosinophils. Research on eosinophils has primarily focused on anti-helminth responses and type 2 immune disorders. In contrast, the search for a role of eosinophils in chronic intestinal inflammation and fibrosis has been limited. With a shift in research focus from adaptive to innate immunity and the fact that the eosinophilic granules are filled with inflammatory mediators, eosinophils are becoming a point of interest in inflammatory bowel diseases. In the current review we summarize eosinophil characteristics and recruitment as well as the current knowledge on presence, inflammatory and pro-fibrotic functions of eosinophils in inflammatory bowel disease and other chronic inflammatory conditions, and we identify research gaps which should be covered in the future.


Eosinophils/physiology , Inflammatory Bowel Diseases/immunology , Models, Immunological , Cell Degranulation , Chemokines/physiology , Chemotaxis, Leukocyte , Eosinophils/chemistry , Fibrosis , Humans , Inflammation , Inflammatory Bowel Diseases/pathology , Interleukins/physiology
11.
Front Immunol ; 12: 757669, 2021.
Article En | MEDLINE | ID: mdl-34603342

The interleukin-6 (IL-6)/IL-12 family of cytokines plays critical roles in the induction and regulation of innate and adaptive immune responses. Among the various cytokines, only this family has the unique characteristic of being composed of two distinct subunits, α- and ß-subunits, which form a heterodimer with subunits that occur in other cytokines as well. Recently, we found a novel intracellular role for one of the α-subunits, Epstein-Barr virus-induced gene 3 (EBI3), in promoting the proper folding of target proteins and augmenting its expression at the protein level by binding to its target protein and a well-characterized lectin chaperone, calnexin, presumably through enhancing chaperone activity. Because calnexin is ubiquitously and constitutively expressed but EBI3 expression is inducible, these results could open an avenue to establish a new paradigm in which EBI3 plays an important role in further increasing the expression of target molecules at the protein level in collaboration with calnexin under inflammatory conditions. This theory well accounts for the heterodimer formation of EBI3 with p28, and probably with p35 and p19 to produce IL-27, IL-35, and IL-39, respectively. In line with this concept, another ß-subunit, p40, plays a critical role in the assembly-induced proper folding of p35 and p19 to produce IL-12 and IL-23, respectively. Thus, chaperone-like activities in proper folding and maturation, which allow the secretion of biologically active heterodimeric cytokines, have recently been highlighted. This review summarizes the current understanding of chaperone-like activities of EBI3 to form heterodimers and other associations together with their possible biological implications.


Calnexin/physiology , Inflammation/metabolism , Interleukins/physiology , Minor Histocompatibility Antigens/physiology , Molecular Chaperones/physiology , Dimerization , Glycoproteins/chemistry , Humans , Interleukins/chemistry , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Neoplasms/metabolism , Neoplasms/pathology , Protein Folding , Protein Interaction Mapping , Protein Subunits , Receptors, Interleukin/chemistry
12.
PLoS One ; 16(8): e0247738, 2021.
Article En | MEDLINE | ID: mdl-34383769

The commensal microbiota regulates susceptibility to enteric pathogens by fine-tuning mucosal innate immune responses, but how susceptibility to enteric viruses is shaped by the microbiota remains incompletely understood. Past reports have indicated that commensal bacteria may either promote or repress rotavirus replication in the small intestine of mice. We now report that rotavirus replicated more efficiently in the intestines of germ-free and antibiotic-treated mice compared to animals with an unmodified microbiota. Antibiotic treatment also facilitated rotavirus replication in type I and type III interferon (IFN) receptor-deficient mice, revealing IFN-independent proviral effects. Expression of interleukin-22 (IL-22) was strongly diminished in the intestine of antibiotic-treated mice. Treatment with exogenous IL-22 blocked rotavirus replication in microbiota-depleted wild-type and Stat1-/- mice, demonstrating that the antiviral effect of IL-22 in animals with altered microbiome is not dependent on IFN signaling. In antibiotic-treated animals, IL-22-induced a specific set of genes including Fut2, encoding fucosyl-transferase 2 that participates in the biosynthesis of fucosylated glycans which can mediate rotavirus binding. Interestingly, IL-22 also blocked rotavirus replication in antibiotic-treated Fut2-/- mice. Furthermore, IL-22 inhibited rotavirus replication in antibiotic-treated mice lacking key molecules of the necroptosis or pyroptosis pathways of programmed cell death. Taken together, our results demonstrate that IL-22 determines rotavirus susceptibility of antibiotic-treated mice, yet the IL-22-induced effector molecules conferring rotavirus resistance remain elusive.


Anti-Bacterial Agents/adverse effects , Interleukins/metabolism , Rotavirus Infections/etiology , Animals , Anti-Bacterial Agents/pharmacology , Disease Susceptibility , Female , Gastrointestinal Microbiome/drug effects , Gene Expression Profiling , Interleukins/physiology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Rotavirus/physiology , Interleukin-22
13.
J Autoimmun ; 123: 102686, 2021 09.
Article En | MEDLINE | ID: mdl-34325305

Cutaneous lupus erythematosus (CLE) is a relapsing autoimmune disease, but key elements that drive disease initiation and progression remain elusive. To date, the lack of ideal murine model which resemble human cutaneous lupus makes it extremely challenging for moving mechanistic discoveries and novel therapeutics. Here, we prompt a humanized murine model to develop an inducible rapid-onset murine that performs cutaneous rather than systemic lupus, depending on the successful human immune system reconstruction from active lupus patients and UVB irradiation as for essentially pathogenic triggers. In addition, we demonstrate a newly discovered population of B cell with a unique phenotype, that of the age-associated B cell (ABC, T-bet+ CD11b+), exhibits B cell clusters in humanized cutaneous lupus. In the response of IL-21 and TLR7/9 signals, recruitment of autoreactive B cells to the position of inflammation with subsequent localized antibody production of IgG2a, IgG2b, IgG3, has the potential to exacerbate ongoing inflammation and thus driving lupus-like autoimmunity in a B-cell-dominant fashion. Overall, our model provides a relevant system for exploring the pathophysiology of cutaneous lupus, a suitable model for drug development, as well as updating a potential role of IL-21 and TLR7/9 to be targeted by biologics.


Aging/immunology , B-Lymphocytes/physiology , Interleukins/physiology , Lupus Erythematosus, Cutaneous/etiology , Adolescent , Adult , Animals , Disease Models, Animal , Female , Humans , Immunoglobulin G/biosynthesis , Lupus Erythematosus, Cutaneous/immunology , Mice , Mice, Inbred BALB C , Middle Aged , Toll-Like Receptor 7/physiology , Toll-Like Receptor 9/physiology , Young Adult
14.
Front Immunol ; 12: 645835, 2021.
Article En | MEDLINE | ID: mdl-34177894

Chronic hepatitis B virus (HBV) infection induces dysfunction of immune response and chronic liver damage. However, the mechanisms that account for HBV-related hepatocellular carcinoma (HCC) are poorly understood. The aim of present study was to investigate the modulatory role of interleukin (IL)-35, an immunosuppressive cytokine, to IL-9-secreting T cells in hepatitis B-related HCC. Twenty-two HBV-related HCC patients, twenty-seven chronic hepatitis B (CHB) patients, and eleven controls were enrolled. Serum IL-35 and IL-9 concentration was measured by ELISA. Peripheral and liver-infiltrating non-specific and HBV-specific Th9 and Tc9 cells were assessed by flow cytometry. The regulatory activity of IL-35 to peripheral and liver-infiltrating Th9 cells was assessed in co-culture system between CD8+ T cells and HepG2.2.15 cells. Serum IL-35 was up-regulated, while IL-9 was down-regulated in HBV-related HCC patients compared with in CHB patients and controls. Peripheral non-specific and HBV-specific Th9 cells, but not Tc9 cells, were decreased in HBV-related HCC patients. Liver-infiltrating non-specific and HBV-specific Th9 cells were also reduced in HCC tumor sites. CD8+ T cells from CHB and HBV-related HCC patients revealed decreased cytotoxicity compared with those from controls. Autologous Th9 cells mediated the elevation of CD8+ T cell cytotoxicity, and this process was depending on IL-9 secretion. Recombinant IL-35 stimulation inhibited IL-9 secretion and PU.1 mRNA expression in non-specific and HBV-specific Th9 cells, leading to the suppression of Th9-mediated CD8+ T cell cytotoxicity in CHB and HBV-related HCC patients. Our current data indicated that IL-35 might dampen non-specific and HBV-specific Th9 cells activity in HBV-related HCC patients.


CD4-Positive T-Lymphocytes/immunology , Carcinoma, Hepatocellular/immunology , Hepatitis B, Chronic/complications , Interleukin-9/biosynthesis , Interleukins/physiology , Liver Neoplasms/immunology , Adult , Aged , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Female , Humans , Interleukin-9/blood , Interleukins/blood , Male , Middle Aged
15.
Front Immunol ; 12: 645717, 2021.
Article En | MEDLINE | ID: mdl-33815402

Idiopathic Pulmonary Fibrosis (IPF) is a severe fibrotic lung disease characterized by excessive collagen deposition and progressive decline in lung function. Th2 T cell-derived cytokines including IL-4 and IL-13 have been shown to contribute to inflammation and fibrotic remodeling in multiple tissues. Interleukin-31 (IL-31) is a newly identified cytokine that is predominantly produced by CD4 Th2 T cells, but its signaling receptor IL-31RA is primarily expressed by non-hematopoietic cells. However, the potential role of the IL-31-IL31RA axis in pulmonary inflammation and fibrosis has remained largely unknown. To determine the role of IL-31RA deficiency in pulmonary fibrosis, wildtype, and IL-31RA knockout mice were treated with bleomycin and measured changes in collagen deposition and lung function. Notably, the loss of IL-31 signaling attenuated collagen deposition and lung function decline during bleomycin-induced pulmonary fibrosis. The total lung transcriptome analysis showed a significant reduction in fibrosis-associated gene transcripts including extracellular matrix and epithelial cell-associated gene networks. Furthermore, the lungs of human IPF showed an elevated expression of IL-31 when compared to healthy subjects. In support, the percentage of IL-31 producing CD4+ T cells was greater in the lungs and PBMCs from IPF patients compared to healthy controls. Our findings suggest a pathogenic role for IL-31/IL-31RA signaling during bleomycin-induced pulmonary fibrosis. Thus, therapeutic targeting the IL-31-IL-31RA axis may prevent collagen deposition, improve lung function, and have therapeutic potential in pulmonary fibrosis.


Idiopathic Pulmonary Fibrosis/drug therapy , Receptors, Interleukin/physiology , Animals , Bleomycin/toxicity , Collagen/metabolism , Female , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/physiopathology , Interleukins/physiology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin/antagonists & inhibitors
16.
J Cell Mol Med ; 25(10): 4721-4731, 2021 05.
Article En | MEDLINE | ID: mdl-33734594

The aryl hydrocarbon receptor (AHR) controls several inflammatory and metabolic pathways involved in various diseases, including the development of arthritis. Here, we investigated the role of AHR activation in IL-22-dependent acute arthritis using the K/BxN serum transfer model. We observed an overall reduction of cytokine expression in Ahr-deficient mice, along with decreased signs of joint inflammation. Conversely, we report worsened arthritis symptoms in Il-22 deficient mice. Pharmacological stimulation of AHR with the agonist VAG539, as well as injection of recombinant IL-22, given prior arthritogenic triggering, attenuated inflammation and reduced joint destruction. The protective effect of VAG539 was abrogated in Il-22 deficient mice. Finally, conditional Ahr depletion of Rorc-expressing cells was sufficient to attenuate arthritis, thereby uncovering a previously unsuspected role of AHR in type 3 innate lymphoid cells during acute arthritis.


Arthritis, Experimental/pathology , Basic Helix-Loop-Helix Transcription Factors/physiology , Immunity, Innate/immunology , Inflammation/pathology , Interleukins/physiology , Joints/pathology , Lymphocytes/pathology , Receptors, Aryl Hydrocarbon/physiology , Acute Disease , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Female , Inflammation/etiology , Inflammation/metabolism , Joints/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Interleukin-22
17.
Life Sci ; 271: 119195, 2021 Apr 15.
Article En | MEDLINE | ID: mdl-33581125

AIMS: Ulcerative colitis and Crohn's disease, collectively known as inflammatory bowel disease (IBD), are chronic inflammatory disorders of the intestine for which key elements in disease initiation and perpetuation are defects in epithelial barrier integrity. Achieving mucosal healing is essential to ameliorate disease outcome and so new therapies leading to epithelial homeostasis and repair are under investigation. This study was designed to determine the mechanisms by which IL-22 regulates intestinal epithelial cell function. MAIN METHODS: Human intestinal organoids and resections, as well as mice were used to evaluate the effect of IL-22 on stem cell expansion, proliferation and expression of mucus components. IL-22 effect on barrier function was assessed in polarized T-84 cell monolayers. Butyrate co-treatments and organoid co-cultures with immune cells were performed to monitor the impact of microbial-derived metabolites and inflammatory environments on IL-22 responses. KEY FINDINGS: IL-22 led to epithelial stem cell expansion, proliferation, barrier dysfunction and anti-microbial peptide production in human and mouse models evaluated. IL-22 also altered the mucus layer by inducing an increase in membrane mucus but a decrease in secreted mucus and goblet cell content. IL-22 had the same effect on anti-microbial peptides and membrane mucus in both healthy and IBD human samples. In contrast, this IL-22-associated epithelial phenotype was different when treatments were performed in presence of butyrate and organoids co-cultured with immune cells. SIGNIFICANCE: Our data indicate that IL-22 promotes epithelial regeneration, innate defense and membrane mucus production, strongly supporting the potential clinical utility of IL-22 as a mucosal healing therapy in IBD.


Epithelial Cells/physiology , Homeostasis/physiology , Interleukins/physiology , Interleukins/therapeutic use , Intestinal Mucosa/physiology , Animals , Cell Line , Coculture Techniques , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Epithelial Cells/drug effects , Homeostasis/drug effects , Humans , Interleukins/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Mice , Mice, Inbred C57BL , Organoids/drug effects , Organoids/physiology , Interleukin-22
18.
Biosci Rep ; 41(1)2021 01 29.
Article En | MEDLINE | ID: mdl-33393621

Psoriasis is a chronic inflammation-associated skin disorder featured by excessive proliferation and abnormal differentiation of keratinocytes. Here, we intended to investigate the role of circular RNA 0061012 (circ_0061012) in psoriasis progression. The expression of circ_0061012, SLMO2-ATP5E readthrough (SLMO2-ATP5E) messenger RNA (mRNA), microRNA-194-5p (miR-194-5p) and GRB2 associated binding protein 1 (GAB1) mRNA was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation and metastasis were analyzed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and transwell assays. Western blot assay was used to measure the protein levels of Ki67, matrix metallopeptidase 9 (MMP9) and GAB1. Dual-luciferase reporter assay and RNA immune co-precipitation (RIP) assay were used to verify the interaction between miR-194-5p and circ_0061012 or GAB1. Circ_0061012 abundance was significantly enhanced in lesional skin samples from psoriasis patients than that in normal skin specimens from healthy volunteers. Interleukin-22 (IL-22) treatment increased the expression of circ_0061012 in a dose-dependent manner. Circ_0061012 silencing alleviated IL-22-induced promoting effects in the proliferation, migration and invasion of HaCaT cells. Circ_0061012 interacted with miR-194-5p, and miR-194-5p knockdown counteracted circ_0061012 silencing-mediated influences in IL-22-induced HaCaT cells. GAB1 was a target of miR-194-5p in HaCaT cells, and miR-194-5p hampered proliferation and metastasis which were induced by IL-22 partly through targeting GAB1. Circ_0061012 elevated the expression of GAB1 through sponging miR-194-5p in HaCaT cells. Circ_0061012 accelerated IL-22-induced proliferation and metastasis in HaCaT cells through enhancing GAB1 expression via sponging miR-194-5p in psoriasis.


Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Interleukins/physiology , Keratinocytes/pathology , MicroRNAs/metabolism , Psoriasis/metabolism , RNA, Circular/physiology , Case-Control Studies , Cell Line , Gene Silencing , Humans , MicroRNAs/genetics , Psoriasis/genetics , Psoriasis/pathology , RNA, Circular/genetics , Interleukin-22
19.
Front Immunol ; 12: 788940, 2021.
Article En | MEDLINE | ID: mdl-34975883

Psoriasis is a complex, chronic relapsing and inflammatory skin disorder with a prevalence of approximately 2% in the general population worldwide. Psoriasis can be triggered by infections, physical injury and certain drugs. The most common type of psoriasis is psoriasis vulgaris, which primarily features dry, well-demarcated, raised red lesions with adherent silvery scales on the skin and joints. Over the past few decades, scientific research has helped us reveal that innate and adaptive immune cells contribute to the chronic inflammatory pathological process of psoriasis. In particular, dysfunctional helper T cells (Th1, Th17, Th22, and Treg cells) are indispensable factors in psoriasis development. When stimulated by certain triggers, antigen-presenting cells (APCs) can release pro-inflammatory factors (IL-23, IFN-α and IL-12), which further activate naive T cells and polarize them into distinct helper T cell subsets that produce numerous cytokines, such as TNF, IFN-γ, IL-17 and IL-22, which act on keratinocytes to amplify psoriatic inflammation. In this review, we describe the function of helper T cells in psoriasis and summarize currently targeted anti-psoriatic therapies.


Psoriasis/immunology , T-Lymphocytes, Helper-Inducer/physiology , Humans , Interleukin-23/antagonists & inhibitors , Interleukins/physiology , Janus Kinase Inhibitors/therapeutic use , Psoriasis/drug therapy , T-Lymphocytes, Regulatory/physiology , Th1 Cells/physiology , Th17 Cells/physiology , Interleukin-22
20.
Mol Neurobiol ; 58(3): 1017-1023, 2021 Mar.
Article En | MEDLINE | ID: mdl-33078369

COVID-19, the global threat to humanity, shares etiological cofactors with multiple diseases including Alzheimer's disease (AD). Understanding the common links between COVID-19 and AD would harness strategizing therapeutic approaches against both. Considering the urgency of formulating COVID-19 medication, its AD association and manifestations have been reviewed here, putting emphasis on memory and learning disruption. COVID-19 and AD share common links with respect to angiotensin-converting enzyme 2 (ACE2) receptors and pro-inflammatory markers such as interleukin-1 (IL-1), IL-6, cytoskeleton-associated protein 4 (CKAP4), galectin-9 (GAL-9 or Gal-9), and APOE4 allele. Common etiological factors and common manifestations described in this review would aid in developing therapeutic strategies for both COVID-19 and AD and thus impact on eradicating the ongoing global threat. Thus, people suffering from COVID-19 or who have come round of it as well as people at risk of developing AD or already suffering from AD, would be benefitted.


Alzheimer Disease/physiopathology , COVID-19/physiopathology , SARS-CoV-2/physiology , Acetylcholine/physiology , Age Factors , Aged , Aged, 80 and over , Alzheimer Disease/complications , Angiotensin-Converting Enzyme 2/physiology , Animals , Anosmia/etiology , Apolipoprotein E4/genetics , Brain/pathology , Brain/virology , COVID-19/complications , Cytokine Release Syndrome/etiology , Cytokines/physiology , Female , Galectins/physiology , Humans , Hypoxia/etiology , Interleukins/physiology , Male , Membrane Proteins/physiology , Mice , Receptors, Virus/physiology , Sex Factors , Smoking/adverse effects
...