Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.901
Filter
1.
Yakugaku Zasshi ; 144(5): 489-496, 2024.
Article in Japanese | MEDLINE | ID: mdl-38692922

ABSTRACT

The tumor necrosis factor receptor (TNFR)-associated factor (TRAF) family of molecules are intracellular adaptors that regulate cellular signaling through members of the TNFR and Toll-like receptor superfamily. Mammals have seven TRAF molecules numbered sequentially from TRAF1 to TRAF7. Although TRAF5 was identified as a potential regulator of TNFR superfamily members, the in vivo function of TRAF5 has not yet been fully elucidated. We identified an unconventional role of TRAF5 in interleukin-6 (IL-6) receptor signaling involving CD4+ T cells. Moreover, TRAF5 binds to the signal-transducing glycoprotein 130 (gp130) receptor for IL-6 and inhibits the activity of the janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway. In addition, Traf5-deficient CD4+ T cells exhibit significantly enhanced IL-6-driven differentiation of T helper 17 (Th17) cells, which exacerbates neuroinflammation in experimental autoimmune encephalomyelitis. Furthermore, TRAF5 demonstrates a similar activity to gp130 for IL-27, another cytokine of the IL-6 family. Additionally, Traf5-deficient CD4+ T cells display significantly increased IL-27-mediated differentiation of Th1 cells, which increases footpad swelling in delayed-type hypersensitivity response. Thus, TRAF5 functions as a negative regulator of gp130 in CD4+ T cells. This review aimed to explain how TRAF5 controls the differentiation of CD4+ T cells and discuss how the expression of TRAF5 in T cells and other cell types can influence the development and progression of autoimmune and inflammatory diseases.


Subject(s)
CD4-Positive T-Lymphocytes , Encephalomyelitis, Autoimmune, Experimental , Signal Transduction , TNF Receptor-Associated Factor 5 , Humans , Animals , TNF Receptor-Associated Factor 5/genetics , TNF Receptor-Associated Factor 5/metabolism , TNF Receptor-Associated Factor 5/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Cytokine Receptor gp130/physiology , Cytokine Receptor gp130/metabolism , Th17 Cells/immunology , Interleukin-6/metabolism , Interleukin-6/physiology , Cell Differentiation , Receptors, Interleukin-6/physiology , Receptors, Interleukin-6/metabolism , Janus Kinases/metabolism , Janus Kinases/physiology , STAT Transcription Factors/physiology , STAT Transcription Factors/metabolism , Mice
2.
Dis Model Mech ; 16(11)2023 11 01.
Article in English | MEDLINE | ID: mdl-37921007

ABSTRACT

Maternal immune activation (MIA) increases the risks for neurodevelopmental disorders in offspring through inflammatory cytokines, including interleukin-6 (IL-6). We therefore aimed to establish a human two-dimensional (2D) in vitro neural model to investigate the effects of IL-6 exposure on neurodevelopment. IL-6 signal transduction requires two receptors: interleukin-6 signal transducer (IL6ST) and interleukin-6 receptor (IL6R). Prenatally, neural cells lack IL6R, and hence cannot elicit cis IL-6 signaling, but IL6R can be provided by microglia in trans. We demonstrate here that an immortalized human neural progenitor cell (NPC) line, ReNCell CX, expresses IL6ST and elicits both cis and trans IL-6 signaling, limiting its use as a model of MIA. In contrast, induced pluripotent stem cell (iPSC)-derived NPCs only activate the IL-6 cascade in trans. Activation of the trans IL-6 cascade did not result in increased proliferation of iPSC-derived NPCs or ReNCell CX, as has been demonstrated in animal models. iPSC-derived NPCs upregulated NR2F1 expression in response to IL-6 signaling in line with analogous experiments in organoids. Thus, iPSC-derived NPCs can be used to model gene expression changes in response to MIA in 2D cultures.


Subject(s)
Induced Pluripotent Stem Cells , Neural Stem Cells , Pluripotent Stem Cells , Animals , Humans , Cell Differentiation , Cytokines/metabolism , Induced Pluripotent Stem Cells/metabolism , Interleukin-6/physiology , Neural Stem Cells/metabolism , Neurons/metabolism , Pluripotent Stem Cells/metabolism
3.
Acta cir. bras ; 38: e381523, 2023. tab, ilus
Article in English | LILACS, VETINDEX | ID: biblio-1439109

ABSTRACT

Purpose: It was aimed to investigate the biochemical and immunohistochemical effects of ephedrine (EPH) in bilateral ovariectomized rats. Methods: Twenty-four Sprague Dawley female rats were divided into three groups: control group: The abdomen was opened and closed without any treatment; ischemia-reperfusion (IR) group: 2 h of ischemia followed by 2 h of reperfusion were allowed to cause IR injury; IR+EPH group: oral EPH solution (5 mg/kg) was administered for 28 days. Results: Biochemical parameters were statistically significant in group comparisons. Increased interleukin-6 (IL-6) expression, degenerative preantral and antral follicle cells and inflammatory cells around blood vessels were seen in IR group. Negative IL-6 expression was observed in seminal epithelial cells, preantral and antral follicle cells in IR+EPH group. While caspase-3 activity increased in granulosa cells and stromal cells in IR group, caspase-3 expression was negative in preantral and antral follicle cells in the germinal epithelium and cortex in IR+EPH group. Conclusion: The effect of apoptosis, which occurs with the signaling that starts in the cell nucleus, caused the cessation of the stimulating effect at the nuclear level after EPH administration, and a decrease in the antioxidative effect in IR damage and inflammation in the apoptotic process.


Subject(s)
Animals , Female , Rats , Ovary/cytology , Interleukin-6/physiology , Ephedrine/analysis , Caspase 3/physiology , Immunohistochemistry , Rats, Sprague-Dawley , Apoptosis
4.
Exerc Sport Sci Rev ; 50(2): 89-96, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34961755

ABSTRACT

Autophagic dysregulation contributes to liver diseases. Although some investigations have examined the effects of endurance and resistance exercise on autophagy activation, potential myokines responsible for skeletal muscle-liver crosstalk are still unknown. Based on experimental studies and bioinformatics, we hypothesized that interleukin 6 (IL-6) and irisin might be key players in the contraction-induced release of molecules that regulate liver autophagic responses.


Subject(s)
Autophagy , Exercise , Fibronectins , Interleukin-6 , Liver , Fibronectins/physiology , Humans , Interleukin-6/physiology , Muscle, Skeletal
5.
FASEB J ; 35(11): e21989, 2021 11.
Article in English | MEDLINE | ID: mdl-34679197

ABSTRACT

Aging exacerbates neointimal formation by reducing apoptosis of vascular smooth muscle cells (VSMCs) and induces inflammation within vascular wall. Prep1 is a homeodomain transcription factor which stimulates the expression of proinflammatory cytokines in aortic endothelial cell models and plays a primary role in the regulation of apoptosis. In this study, we have investigated the role of Prep1 in aorta of Prep1 hypomorphic heterozygous mice (Prep1i/+ ) and in VSMCs, and its correlation with aging. Histological analysis from Prep1i/+ aortas revealed a 25% reduction in medial smooth muscle cell density compared to WT animals. This result paralleled higher apoptosis, caspase 3, caspase 9 and p53 levels in Prep1i/+ mice and lower Bcl-xL. Prep1 overexpression in VSMCs decreased apoptosis by 25% and caspase 3 and caspase 9 expression by 40% and 37%. In parallel, Bcl-xL inhibition by BH3I-1 and p53 induction by etoposide reverted the antiapoptotic effect of Prep1. Experiments performed in aorta from 18 months old WT mice showed a significant increase in Prep1, p16INK4 , p21Waf1 and interleukin 6 (IL-6) compared to youngest animals. Similar results have been observed in H2 O2 -induced senescent VSMCs. Interestingly, the synthetic Prep1 inhibitory peptide Prep1 (54-72) reduced the antiapoptotic effects mediated by IL-6, particularly in senescent VSMCs. These results indicate that IL-6-Prep1 signaling reduces apoptosis, by modulating Bcl-xL and p53 both in murine aorta and in VSMCs. In addition, age-dependent increase in IL-6 and Prep1 in senescent VSMCs and in old mice may be involved in the aging-related vascular dysfunction.


Subject(s)
Aging/metabolism , Homeodomain Proteins/physiology , Interleukin-6/physiology , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Apoptosis , Cells, Cultured , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism
6.
Biol Psychol ; 165: 108147, 2021 10.
Article in English | MEDLINE | ID: mdl-34492333

ABSTRACT

Although elevations in systemic suPAR levels have been associated with inflammatory conditions and with exposure to life stress and adversity, it is not yet clear whether acute psychological stress influences suPAR levels, either systemically and/or in saliva. The aim of this study was to investigate whether salivary suPAR levels are increased following exposure to acute psychological stress. Healthy subjects, aged 18-40 years, completed a laboratory psychological stressor and provided saliva samples before and after the stress test (60 min apart). Levels of suPAR as well as those of cytokines increased in the post-stress samples (all ps < .001). Baseline and post-stress IL-1ß and TNF-α as well as post-stress IL-6 correlated significantly with suPAR (all ps < .01), but IL-10 and baseline IL-6 did not. These results show that suPAR levels in saliva are stress-reactive and suggest a potential application as stress biomarkers in saliva, particularly given the advantage of easily detectable concentrations.


Subject(s)
Receptors, Urokinase Plasminogen Activator , Saliva , Stress, Psychological , Biomarkers , Humans , Interleukin-10/blood , Interleukin-10/physiology , Interleukin-1beta/blood , Interleukin-1beta/physiology , Interleukin-6/blood , Interleukin-6/physiology , Receptors, Urokinase Plasminogen Activator/blood , Receptors, Urokinase Plasminogen Activator/physiology , Tumor Necrosis Factor-alpha/blood
7.
Am J Nephrol ; 52(9): 745-752, 2021.
Article in English | MEDLINE | ID: mdl-34535589

ABSTRACT

INTRODUCTION: Inflammation is important in the pathogenesis of atherosclerosis. Elevated interleukin-6 (IL-6) is associated with cardiovascular events and also predicts mortality in individuals with CKD. Our goal was to determine the association between IL-6, FGF23, and high-sensitivity C-reactive protein (hsCRP) on coronary artery calcification (CAC) progression and mortality in incident dialysis patients without prior coronary events. METHODS: A prospective cohort of incident adult dialysis participants had CAC measured by ECG-triggered multislice CT scans at baseline and at least 12 months later. Lipids, mineral metabolism markers, FGF23, and inflammatory markers, such as IL-6 and hsCRP, were measured at the baseline visit. RESULTS: Participants in the high IL-6 tertile had the highest baseline CAC score (133.25 [10.35-466.15]) compared to the low (0.25 [0-212.2]) and intermediate (29.55 [0-182.85]) tertiles. Almost half of the participants with high IL-6 (15 of 32 [46.9%]) experienced progression of CAC compared to participants with low (8 of 32 [25%]) and intermediate (9 of 32 [28.1%]) (p = 0.05) IL-6 levels. Each log increase in IL-6 was associated with increase in death (hazard ratio 2.2, 95% CI: 1.2-3.8; p = 0.01). After adjusting for smoking, age, gender, race, diabetes, phosphate, and baseline calcium score, IL-6 (log) was associated with 2.2 times (95% CI: 1.1-4.6; p = 0.03) increase in death. CONCLUSION: IL-6 is associated with progression of CAC and mortality in incident dialysis patients.


Subject(s)
C-Reactive Protein/physiology , Coronary Artery Disease/etiology , Coronary Artery Disease/mortality , Interleukin-6/physiology , Renal Dialysis , Vascular Calcification/etiology , Vascular Calcification/mortality , Adult , Cohort Studies , Disease Progression , Female , Humans , Male , Middle Aged , Prospective Studies
8.
Front Immunol ; 12: 711847, 2021.
Article in English | MEDLINE | ID: mdl-34484214

ABSTRACT

Severe fever with thrombocytopenia syndrome (SFTS) is a new tick-borne viral disease, and most SFTS virus (SFTSV) infections occur via bites from the tick Haemaphysalis longicornis; however, SFTSV transmission can also occur through close contact with an infected patient. SFTS is characterized by acute high fever, thrombocytopenia, leukopenia, elevated serum hepatic enzyme levels, gastrointestinal symptoms, and multiorgan failure and has a 16.2 to 30% mortality rate. In this study, we found that age, dyspnea rates, aspartate aminotransferase (AST), alanine aminotransferase (ALT), lactate dehydrogenase, multiorgan dysfunction score (MODS), viral load, IL-6 levels, and IL-10 levels were higher in patients with fatal disease than in patients with nonfatal disease during the initial clinical course of SFTS. In addition, we found that IL-6 and IL-10 levels, rather than viral load and neutralizing antibody titers, in patients with an SFTSV infection strongly correlated with outcomes (for severe disease with an ultimate outcome of recovery or death).


Subject(s)
Interleukin-10/blood , Interleukin-6/blood , Severe Fever with Thrombocytopenia Syndrome/immunology , Viremia/immunology , Aged , Aged, 80 and over , Alanine Transaminase/blood , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Aspartate Aminotransferases/blood , Cytokines/blood , Dyspnea/etiology , Female , Humans , Interleukin-10/physiology , Interleukin-6/physiology , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Multiple Organ Failure/etiology , Multiple Organ Failure/mortality , Phlebovirus/immunology , Republic of Korea/epidemiology , Severe Fever with Thrombocytopenia Syndrome/blood , Severe Fever with Thrombocytopenia Syndrome/mortality , Severe Fever with Thrombocytopenia Syndrome/virology , Treatment Outcome , Viral Load , Viremia/blood , Viremia/mortality
9.
Front Immunol ; 12: 675542, 2021.
Article in English | MEDLINE | ID: mdl-34394075

ABSTRACT

Autoreactive T cells play a crucial role in the pathogenesis of systemic lupus erythematosus (SLE). TGF-ß type I receptor (TGFßRI) is pivotal in determining T cell activation. Here, we showed that TGFßRI expression in naïve CD4+ T cells was decreased in SLE patients, especially in those with high disease activity. Moreover, IL-6 was found to downregulate TGFßRI expression through JAK/STAT3 pathway in SLE patients. In vitro, the JAK inhibitor tofacitinib inhibited SLE T cell activating by upregulating TGFßRI expression in a dose-dependent manner. In MRL/lpr mice, tofacitinib treatment ameliorated the clinical indicators and lupus nephritis, as evidenced by reduced plasma anti-dsDNA antibody levels, decreased proteinuria, and lower renal histopathological score. Consistently, tofacitinib enhanced TGFßRI expression and inhibited T cell activation in vivo. TGFßRI inhibitor SB431542 reversed the effects of tofacitinib on T cell activation. Thus, our results have indicated that tofacitinib can suppress T cell activation by upregulating TGFßRI expression, which provides a possible molecular mechanism underlying clinical efficacy of tofacitinib in treating SLE patients.


Subject(s)
Lupus Erythematosus, Systemic/drug therapy , Lymphocyte Activation/drug effects , Piperidines/pharmacology , Pyrimidines/pharmacology , Receptor, Transforming Growth Factor-beta Type I/physiology , T-Lymphocytes/drug effects , Adult , Animals , Female , Humans , Interleukin-6/physiology , Janus Kinases/physiology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Middle Aged , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/genetics , STAT3 Transcription Factor/physiology , T-Lymphocytes/immunology , Young Adult
10.
Immunol Lett ; 237: 58-65, 2021 09.
Article in English | MEDLINE | ID: mdl-34246712

ABSTRACT

Type 2 Diabetes is a chronic disease resulting from insulin dysfunction that triggers a low-grade inflammatory state and immune impairment. Leishmaniasis is an infectious disease characterized by chronic inflammation resulted from the parasite's immunomodulation ability. Thus, due to the delicate immune balance required in the combat and resistance to Leishmania infection and the chronic deregulation of the inflammatory response observed in type 2 diabetes, we evaluated the response of PBMC from diabetic patients to in vitro Leishmania amazonensis infection. For that, peripheral blood was collected from 25 diabetic patients and 25 healthy controls matched for age for cells extraction and subsequent experimental infection for 2 or 24 h and analyzed for phagocytic and leishmanicidal capacity by optical microscopy, oxidative stress by GSSG generation, labeling of intracellular mediators by enzyme-Linked immunosorbent assay, and cytokines measurement with cytometric beads array technique. We found that the diabetic group had a higher percentage of infected cells and a greater number of amastigotes per cell. Also, even inducing NF-kB phosphorylation and increasing TNF production after infection, cells from diabetic patients were unable to downregulate NRF2 and generate oxidative stress, which may be associated with the exacerbated levels of IL-6 observed. PBMC of diabetic individuals are more susceptible to infection by L. amazonensis and fail to control the infection over time due to the inability to generate effector microbicidal molecules.


Subject(s)
Cytokines/physiology , Diabetes Mellitus, Type 2/immunology , Leishmania mexicana/pathogenicity , Leishmaniasis, Cutaneous/etiology , Leukocytes, Mononuclear/parasitology , NF-E2-Related Factor 2/deficiency , Aged , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Disease Susceptibility , Female , Glutathione/blood , Glycated Hemoglobin/analysis , Humans , Immunocompetence , In Vitro Techniques , Inflammation , Interleukin-6/physiology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/parasitology , Male , Middle Aged , NF-E2-Related Factor 2/physiology , Nitric Oxide/metabolism , Oxidative Stress , Respiratory Burst , Tumor Necrosis Factor-alpha/physiology
11.
Psychoneuroendocrinology ; 131: 105284, 2021 09.
Article in English | MEDLINE | ID: mdl-34090139

ABSTRACT

The pro-inflammatory role of interleukin-6 (IL-6) is well-characterized. Blockade of IL-6, by Tocilizumab, is used in patients with rheumatoid arthritis and those diagnosed with cytokine storm. However, brain-produced IL-6 has recently emerged as a critical mediator of gut/adipose communication with the brain. Central nervous system (CNS) IL-6 is engaged by peripheral and central signals regulating energy homeostasis. IL-6 is critical for mediating hypophagia and weight loss effects of a GLP-1 analog, exendin-4, a clinically utilized drug. However, neuroanatomical substrates and behavioral mechanisms of brain IL-6 energy balance control remain poorly understood. We propose that the lateral hypothalamus (LH) is an IL-6-harboring brain region, key to food intake and food reward control. Microinjections of IL-6 into the LH reduced chow and palatable food intake in male rats. In contrast, female rats responded with reduced motivated behavior for sucrose, measured by the progressive ratio operant conditioning test, a behavioral mechanism previously not linked to IL-6. To test whether IL-6, produced in the LH, is necessary for ingestive and motivated behaviors, and body weight homeostasis, virogenetic knockdown by infusion of AAV-siRNA-IL6 into the LH was utilized. Attenuation of LH IL-6 resulted in a potent increase in sucrose-motivated behavior, without any effect on ingestive behavior or body weight in female rats. In contrast, the treatment did not affect any parameters measured (chow intake, sucrose-motivated behavior, locomotion, and body weight) in chow-fed males. However, when challenged with a high-fat/high-sugar diet, the male LH IL-6 knockdown rats displayed rapid weight gain and hyperphagia. Together, our data suggest that LH-produced IL-6 is necessary and sufficient for ingestive behavior and weight homeostasis in male rats. In females, IL-6 in the LH plays a critical role in food-motivated, but not ingestive behavior control or weight regulation. Thus, collectively these data support the idea that brain-produced IL-6 engages the hypothalamus to control feeding behavior.


Subject(s)
Body Weight , Feeding Behavior , Hypothalamus , Interleukin-6 , Motivation , Animals , Body Weight/physiology , Feeding Behavior/physiology , Female , Hypothalamus/metabolism , Interleukin-6/physiology , Male , Motivation/physiology , Rats
12.
Psychoneuroendocrinology ; 131: 105295, 2021 09.
Article in English | MEDLINE | ID: mdl-34119855

ABSTRACT

The majority of COVID-19 survivors experience long-term neuropsychiatric symptoms such as fatigue, sleeping difficulties, depression and anxiety. We propose that neuroimmune cross-talk via inflammatory cytokines such as interleukin-6 (IL-6) could underpin these long-term COVID-19 symptoms. This hypothesis is supported by several lines of research, including population-based cohort and genetic Mendelian Randomisation studies suggesting that inflammation is associated with fatigue and sleeping difficulties, and that IL-6 could represent a possible causal driver for these symptoms. Immune activation following COVID-19 can disrupt T helper 17 (TH17) and regulatory T (Treg) cell responses, affect central learning and emotional processes, and lead to a vicious cycle of inflammation and mitochondrial dysfunction that amplifies the inflammatory process and results in immuno-metabolic constraints on neuronal energy metabolism, with fatigue being the ultimate result. Increased cytokine activity drives this process and could be targeted to interrupt it. Therefore, whether persistent IL-6 dysregulation contributes to COVID-19-related long-term fatigue, sleeping difficulties, depression, and anxiety, and whether targeting IL-6 pathways could be helpful for treatment and prevention of long COVID are important questions that require investigation. This line of research could inform new approaches for treatment and prevention of long-term neuropsychiatric symptoms of COVID-19. Effective treatment and prevention of this condition could also help to stem the anticipated rise in depression and other mental illnesses ensuing this pandemic.


Subject(s)
COVID-19/complications , Interleukin-6/physiology , Mental Disorders/etiology , Animals , Anxiety/epidemiology , Anxiety/etiology , COVID-19/epidemiology , COVID-19/etiology , COVID-19/psychology , Cohort Studies , Depression/epidemiology , Depression/etiology , Fatigue/epidemiology , Fatigue/etiology , Humans , Interleukin-6/metabolism , Interleukin-6/pharmacology , Mental Disorders/epidemiology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , SARS-CoV-2/physiology , Sleep Wake Disorders/epidemiology , Sleep Wake Disorders/etiology , Survivors/statistics & numerical data , Post-Acute COVID-19 Syndrome
13.
Expert Rev Clin Pharmacol ; 14(10): 1279-1287, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34187281

ABSTRACT

Introduction: Tocilizumab is one of the main repurposed therapies investigated for COVID-19 pneumonia since the start of the pandemic, but there has been conflicting evidence for its use.Areas covered: This review covers the physiology of interleukin-6 and its role in the pathophysiology of COVID-19. We discuss the use of tocilizumab in other diseases and the rationale for its use in COVID-19. We summarize the design, contrasting results, and implications of the clinical trials of tocilizumab in COVID-19 to date and discuss the current guidance for its use.Expert opinion: The evidence to date suggests benefit with the use of tocilizumab in some but not all patients with COVID-19. Benefit seems to be greatest when given early after clinical deterioration with the presence of systemic inflammation. However, questions remain around the optimal timing, patient selection, and concomitant treatments.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Interleukin-6/physiology , SARS-CoV-2 , COVID-19/physiopathology , Humans
14.
J Invest Dermatol ; 141(11): 2630-2638.e7, 2021 11.
Article in English | MEDLINE | ID: mdl-34029573

ABSTRACT

Functional studies to delineate the molecular mechanisms of causal genetic variants are the main focus in the post-GWAS era. Previous GWASs have identified >50 susceptibility loci associated with psoriasis. Functional understanding of the biology underlying the disease risk of most of these associated loci is unclear. In this study, we identified a regulatory SNP at the putative enhancer of the LCE3A gene within the epidermal differentiation complex that showed epistatic interaction with HLA-Cw6. The variant allele disrupted signal transducer and activator of transcription 3 binding to the region, thereby regulating the expression of the downstream LCE3A gene. Electrophoretic mobility shift and pulldown assay confirmed the preferential binding of signal transducer and activator of transcription 3 to the DNA with a wild-type allele compared with the DNA with a variant allele. The reporter assay further validated the IL-6‒stimulated phosphorylated signal transducer and activator of transcription 3‒mediated LCE3A activation in the presence of the wild-type allele. Interestingly, the presence of the HLA-Cw6 allele leads to IL-6‒mediated phosphorylation of signal transducer and activator of transcription 3, followed by its nuclear localization in the epidermal keratinocytes of psoriatic skin, suggesting indirect interaction of the HLA-Cw6 allele and a regulatory SNP upstream of the LCE3A gene. This study reflects an interesting approach to dissecting the molecular mechanism underlying the genetic interaction observed between HLA-Cw6 and LCE3A in psoriasis pathogenesis.


Subject(s)
Cornified Envelope Proline-Rich Proteins/genetics , HLA-C Antigens/genetics , Psoriasis/genetics , Cells, Cultured , Gene Expression Regulation , Humans , Interleukin-1alpha/physiology , Interleukin-6/physiology , Phosphorylation , Polymorphism, Single Nucleotide , Psoriasis/etiology , STAT3 Transcription Factor/metabolism
15.
J Hepatol ; 75(3): 647-658, 2021 09.
Article in English | MEDLINE | ID: mdl-33991637

ABSTRACT

BACKGROUND AND AIMS: COVID-19 is associated with liver injury and elevated interleukin-6 (IL-6). We hypothesized that IL-6 trans-signaling in liver sinusoidal endothelial cells (LSECs) leads to endotheliopathy (a proinflammatory and procoagulant state) and liver injury in COVID-19. METHODS: Coagulopathy, endotheliopathy, and alanine aminotransferase (ALT) were retrospectively analyzed in a subset (n = 68), followed by a larger cohort (n = 3,780) of patients with COVID-19. Liver histology from 43 patients with COVID-19 was analyzed for endotheliopathy and its relationship to liver injury. Primary human LSECs were used to establish the IL-6 trans-signaling mechanism. RESULTS: Factor VIII, fibrinogen, D-dimer, von Willebrand factor (vWF) activity/antigen (biomarkers of coagulopathy/endotheliopathy) were significantly elevated in patients with COVID-19 and liver injury (elevated ALT). IL-6 positively correlated with vWF antigen (p = 0.02), factor VIII activity (p = 0.02), and D-dimer (p <0.0001). On liver histology, patients with COVID-19 and elevated ALT had significantly increased vWF and platelet staining, supporting a link between liver injury, coagulopathy, and endotheliopathy. Intralobular neutrophils positively correlated with platelet (p <0.0001) and vWF (p <0.01) staining, and IL-6 levels positively correlated with vWF staining (p <0.01). IL-6 trans-signaling leads to increased expression of procoagulant (factor VIII, vWF) and proinflammatory factors, increased cell surface vWF (p <0.01), and increased platelet attachment in LSECs. These effects were blocked by soluble glycoprotein 130 (IL-6 trans-signaling inhibitor), the JAK inhibitor ruxolitinib, and STAT1/3 small-interfering RNA knockdown. Hepatocyte fibrinogen expression was increased by the supernatant of LSECs subjected to IL-6 trans-signaling. CONCLUSION: IL-6 trans-signaling drives the coagulopathy and hepatic endotheliopathy associated with COVID-19 and could be a possible mechanism behind liver injury in these patients. LAY SUMMARY: Patients with SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) infection often have liver injury, but why this occurs remains unknown. High levels of interleukin-6 (IL-6) and its circulating receptor, which form a complex to induce inflammatory signals, have been observed in patients with COVID-19. This paper demonstrates that the IL-6 signaling complex causes harmful changes to liver sinusoidal endothelial cells and may promote blood clotting and contribute to liver injury.


Subject(s)
COVID-19/complications , Endothelial Cells/pathology , Interleukin-6/physiology , Liver Diseases/etiology , SARS-CoV-2 , Adult , Blood Coagulation Disorders/etiology , Fibrinogen/analysis , Humans , Interleukin-6/blood , Janus Kinase 1/metabolism , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Retrospective Studies , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , von Willebrand Factor/analysis
16.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802761

ABSTRACT

Among patients suffering from coronavirus disease 2019 (COVID-19) syndrome, one of the worst possible scenarios is represented by the critical lung damage caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-induced cytokine storm, responsible for a potentially very dangerous hyperinflammatory condition. Within such a context, interleukin-6 (IL-6) plays a key pathogenic role, thus being a suitable therapeutic target. Indeed, the IL-6-receptor antagonist tocilizumab, already approved for treatment of refractory rheumatoid arthritis, is often used to treat patients with severe COVID-19 symptoms and lung involvement. Therefore, the aim of this review article is to focus on the rationale of tocilizumab utilization in the SARS-CoV-2-triggered cytokine storm, as well as to discuss current evidence and future perspectives, especially with regard to ongoing trials referring to the evaluation of tocilizumab's therapeutic effects in patients with life-threatening SARS-CoV-2 infection.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , SARS-CoV-2/immunology , Antibodies, Monoclonal, Humanized/chemistry , COVID-19/complications , COVID-19/immunology , COVID-19/physiopathology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/physiopathology , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/physiology
17.
Biochim Biophys Acta Mol Cell Res ; 1868(7): 119037, 2021 06.
Article in English | MEDLINE | ID: mdl-33839168

ABSTRACT

Interleukin-6 (IL-6) enhanced TNF-α and TRAIL/Apo2L induced cell death in various human cancer cells derived from malignant glioma, melanoma, breast cancer and leukemia, although the effect was not detected with IL-6 alone. The effects of IL-6 using SKBR3 cells were associated with the generation of apoptotic cells as analyzed by fluorescence microscopy and flow cytometry. IL-6 activated p53 and upregulated TRAIL death receptors (DR-4 and DR-5) and stimulated the TNF-α and TRAIL dependent extrinsic apoptotic pathway without activation of the p53 mediated intrinsic apoptotic pathway. TNF-α and TRAIL induced cleavage of caspase-8 and caspase-3 was more enhanced by IL-6, although these caspases were not cleaved by IL-6 alone. The dead cell generation elicited by the combination with IL-6 was blocked by anti-human TRAIL R2/TNFRSF10B Fc chimera antibody which can neutralize the DR-5 mediated death signal. These findings indicate that IL-6 could contribute to the enhancement of TNF-α or TRAIL induced apoptosis through p53 dependent upregulation of DR-4 and DR-5. The data suggest that a favorable therapeutic interaction could occur between TNF-α or TRAIL and IL-6, and provide an experimental basis for rational clinical treatments in various cancers.


Subject(s)
Interleukin-6/metabolism , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Caspases/metabolism , Cell Death/physiology , Cell Line, Tumor/metabolism , Humans , Interleukin-6/physiology , Neoplasms/physiopathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Death Domain/metabolism , Receptors, Death Domain/physiology , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Front Immunol ; 12: 630196, 2021.
Article in English | MEDLINE | ID: mdl-33897686

ABSTRACT

Objective: Kawasaki disease (KD) is the most common cause of acquired pediatric heart disease in the developed world. 10% of KD patients are resistant to front-line therapy, and no interventions exist to address secondary complications such as myocardial fibrosis. We sought to identify proteins and pathways associated with disease and anti-IL-1 treatment in a mouse model of KD. Methods: Vasculitis was induced via Lactobacillus casei cell wall extract (LCWE) injection in 5-week-old male mice. Groups of mice were injected with LCWE alone, LCWE and IL-1 receptor antagonist anakinra, or saline for controls. Upper heart tissue was assessed by quantitative mass spectrometry analysis. Expression and activation of STAT3 was assessed by immunohistochemistry, immunofluorescence and Western blot, and IL-6 expression by RNA-seq and ELISA. A STAT3 small molecular inhibitor and anti-IL-6R antibody were used to evaluate the role of STAT3 and IL-6 in disease development. Results: STAT3 was highly expressed and phosphorylated in cardiac tissue of LCWE-injected mice, and reduced following anakinra treatment. Il6 and Stat3 gene expression was enhanced in abdominal aorta of LCWE-injected mice and reduced with Anakinra treatment. IL-6 serum levels were enhanced in LCWE-injected mice and normalized by anakinra. However, neither inhibition of STAT3 nor blockade of IL-6 altered disease development. Conclusion: Proteomic analysis of cardiac tissues demonstrates differential protein expression between KD-like, control and anakinra treated cardiac tissue. STAT3 and IL-6 were highly upregulated with LCWE and normalized by anakinra treatment. However, both STAT3 and IL-6 were dispensable for disease development indicating they may be bystanders of inflammation.


Subject(s)
Interleukin-6/physiology , Mucocutaneous Lymph Node Syndrome/etiology , STAT3 Transcription Factor/physiology , Serum Amyloid A Protein/antagonists & inhibitors , Animals , Cell Wall , Disease Models, Animal , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-6/antagonists & inhibitors , Interleukin-6/blood , Lacticaseibacillus casei , Male , Mice , Mice, Inbred C57BL , Mucocutaneous Lymph Node Syndrome/drug therapy , Myocardium/metabolism , Proteomics , STAT3 Transcription Factor/analysis , STAT3 Transcription Factor/antagonists & inhibitors
19.
Cell Host Microbe ; 29(6): 959-974.e7, 2021 06 09.
Article in English | MEDLINE | ID: mdl-33894128

ABSTRACT

Microbiota play critical roles in regulating colitis and colorectal cancer (CRC). However, it is unclear how the microbiota generate protective immunity against these disease states. Here, we find that loss of the innate and adaptive immune signaling molecule, TAK1, in myeloid cells (Tak1ΔM/ΔM) yields complete resistance to chemical-induced colitis and CRC through microbiome alterations that drive protective immunity. Tak1ΔM/ΔM mice exhibit altered microbiota that are critical for resistance, with antibiotic-mediated disruption ablating protection and Tak1ΔM/ΔM microbiota transfer conferring protection against colitis or CRC. The altered microbiota of Tak1ΔM/ΔM mice promote IL-1ß and IL-6 signaling pathways, which are required for induction of protective intestinal Th17 cells and resistance. Specifically, Odoribacter splanchnicus is abundant in Tak1ΔM/ΔM mice and sufficient to induce intestinal Th17 cell development and confer resistance against colitis and CRC in wild-type mice. These findings identify specific microbiota strains and immune mechanisms that protect against colitis and CRC.


Subject(s)
Bacteroidetes/metabolism , Colitis/microbiology , Colorectal Neoplasms/microbiology , Cytokines/physiology , Gastrointestinal Microbiome , MAP Kinase Kinase Kinases/physiology , Th17 Cells/metabolism , Animals , Colitis/chemically induced , Colitis/metabolism , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Disease Models, Animal , Feces/microbiology , Female , Host Microbial Interactions , Immunity, Innate , Interleukin-1beta/physiology , Interleukin-6/physiology , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Signal Transduction , Th17 Cells/immunology
20.
J Addict Dis ; 39(3): 347-356, 2021.
Article in English | MEDLINE | ID: mdl-33719920

ABSTRACT

BACKGROUND: Opioid use disorder (OUD) is one of the problems and concerns of all countries in the world. On the other hand, transcranial direct current stimulation (tDCS) has been used as a new therapeutic intervention in various psychiatric disorders. OBJECTIVE: This study aimed to investigate the effect of bilateral tDCS on the expression levels of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), craving and impulsive behaviors of male patients with OUD. METHODS: This is a double-blind sham-controlled clinical trial. Participants were 31 male patients with OUD divided into three groups of left anode/right cathode tDCS, right anode/left cathode tDCS, and sham tDCS. They received active tDCS (2 mA, 20 min), applied over their dorsolateral prefrontal cortex (DLPFC) for 10 consecutive days. Expression levels of IL-6 and TNF-α cytokines were measured using ELISA method, and the Desires for Drug Questionnaire and the Barratt Impulsiveness Scale version 11 were used to assess the craving and impulsivity of subjects, respectively. RESULTS: Both active and sham tDCS could significantly reduce drug craving in subjects (p < 0.05). Active tDCS over the right/left DLPFC significantly reduced impulsivity and its dimensions (overall, attentional, motor, and nonplanning) compared to the sham tDCS (p < 0.05). It could also reduce the expression levels of IL-6 and TNF-α, but the difference was not statistically significant. CONCLUSIONS: The active tDCS over the right/left DLPFC, as a noninvasive and complementary treatment, can be used along with other common methods for the treatment of patients with OUD. It can improve their cognitive functions by reducing impulsivity.


Subject(s)
Craving , Impulsive Behavior , Interleukin-6/physiology , Opioid-Related Disorders/therapy , Transcranial Direct Current Stimulation/methods , Tumor Necrosis Factor-alpha/physiology , Adult , Double-Blind Method , Humans , Iran/epidemiology , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...