Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.709
Filter
1.
Steroids ; 211: 109503, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39208922

ABSTRACT

Anabolic-androgenic steroids (AAS) abuse is linked to some abnormalities in several tissues including the kidney. However, the precise molecular mediators involved in AAS-induced kidney disorder remain elusive. The main objective of the present study was to investigate the effect of Nandrolone decanoate on kidney injury alone or in combination with moderate exercise and its related mechanisms. Thirty-two male Wistar rats were subdivided randomly into four groups. control (Con), Nandrolone (10 mg/kg)(N), Exercise (Exe), Nandrolone + Exercise (N+Exe). RESULTS: After 6 weeks, nandrolone treatment led to a significant increase in functional parameters such as serum cystatin c, urea, creatinine, albuminuria and Albumin/ creatinine ratio indicating kidney dysfunction. Moreover, nandrolone treatment increased vacuolization, focal inflammation, hemorragia, cast formation fibrosis in the renal tissue of rats. miRNA-146a increased in kidney tissue after nandrolone exposure by using RT-PCR which may be considered idealtheranomiRNAcandidates for diagnosis and treatment. Western blotting indicated that IRAK1, TRAF6, TNF-α, NF-κB, iNOS and TGF-ß protein expressions were considerably elevated in the kidneys of nandrolone treated rats. Moderate exercise could alleviate the renal dysfunction, histological abnormalities and aforementioned proteins. Our findings suggested that nandrolone consumption can cause damage to kidney tissue probably through miRNA-146a targeting IRAK1 and TRAF6 via activation of the NF-κB and TGF-ß pathway. These results provide future lines of research in the identification of theranoMiRNAs related to nandrolone treatment, which can be ameliorated by moderate exercise.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , MicroRNAs , NF-kappa B , Nandrolone Decanoate , Physical Conditioning, Animal , Rats, Wistar , TNF Receptor-Associated Factor 6 , Animals , Male , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Rats , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Nandrolone/pharmacology , Nandrolone/adverse effects , Signal Transduction/drug effects , Kidney/metabolism , Kidney/drug effects , Kidney/pathology
2.
Cell Rep ; 43(8): 114570, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39093697

ABSTRACT

A wide variety of electrophilic derivatives of itaconate, the Kreb's cycle-derived metabolite, are immunomodulatory, yet these derivatives have overlapping and sometimes contradictory activities. Therefore, we generated a genetic system to interrogate the immunomodulatory functions of endogenously produced itaconate in human macrophages. Endogenous itaconate is driven by multiple innate signals restraining inflammatory cytokine production. Endogenous itaconate directly targets cysteine 13 in IRAK4 (disrupting IRAK4 autophosphorylation and activation), drives the degradation of nuclear factor κB, and modulates global ubiquitination patterns. As a result, cells unable to make itaconate overproduce inflammatory cytokines such as tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), and IL-1ß in response to these innate activators. In contrast, the production of interferon (IFN)ß, downstream of LPS, requires the production of itaconate. These data demonstrate that itaconate is a critical arbiter of inflammatory cytokine production downstream of multiple innate signaling pathways, laying the groundwork for the development of itaconate mimetics for the treatment of autoimmunity.


Subject(s)
Cytokines , Immunity, Innate , Macrophages , Succinates , Ubiquitination , Humans , Succinates/pharmacology , Succinates/metabolism , Ubiquitination/drug effects , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Cytokines/metabolism , Immunity, Innate/drug effects , NF-kappa B/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Signal Transduction/drug effects , Lipopolysaccharides/pharmacology , HEK293 Cells
3.
J Autoimmun ; 148: 103300, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39116634

ABSTRACT

The mechanisms of endotoxin tolerance (ET), which down-regulate inflammation, are well described in response to exogenous toll-like receptor ligands, but few studies have focused on ET-associated mechanisms in inflammatory disease. As blocking TNF can attenuate the development of ET, the effect of anti-TNF on the expression of key ET-associated molecules in inflammatory auto-immune disease was measured; changes in inflammatory gene expression were confirmed using an ET bioassay. The expression of immunomodulatory molecules was measured in a murine model of arthritis treated with anti-TNF and the expression of ET-associated molecules was measured in whole blood in rheumatoid arthritis (RA) and ankylosing spondylitis (AS) patients, before and after therapy. The expression of ET-associated genes was also measured in RA patient monocytes before and after therapy, in anti-TNF responders and non-responders. Tnfaip3, Ptpn6 and Irak3 were differentially expressed in affected paws, spleens, lymph nodes and circulating leucocytes in experimental murine arthritis treated with anti-TNF. Prior to therapy, the expression of TNFAIP3, INPP5D, PTPN6, CD38 and SIGIRR in whole blood differed between human healthy controls and RA or AS patients. In blood monocytes from RA patients, the expression of TNFAIP3 was significantly reduced by anti-TNF therapy in non-responders. Prior to therapy, anti-TNF non-responders had higher expression of TNFAIP3 and SLPI, compared to responders. Although the expression of TNFAIP3 was significantly higher in RA non-responders prior to treatment, the post-treatment reduction to a level similar to responders did not coincide with a clinical response to therapy.


Subject(s)
Arthritis, Rheumatoid , Endotoxins , Immune Tolerance , Spondylitis, Ankylosing , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Immune Tolerance/drug effects , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Endotoxins/immunology , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Female , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Arthritis, Experimental/immunology , Arthritis, Experimental/drug therapy , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Monocytes/immunology , Monocytes/metabolism , Monocytes/drug effects , Middle Aged , Adult , Inflammation/immunology , Disease Models, Animal
4.
Immunobiology ; 229(5): 152835, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38986278

ABSTRACT

Podocytes maintain renal filtration integrity when the glomerular filtration barrier (GFB) is integrated. Impairment or attrition of podocytes, leading to compromised GFB permeability, constitutes the primary etiology of proteinuria and is a hallmark pathological feature of diabetic nephropathy (DN). This study centers on Heterogeneous Nuclear Ribonucleoprotein I (HNRNP I), an RNA-binding protein, delineating its role in facilitating DN-induced renal damage by modulating podocyte health. Comparative analyses in renal biopsy specimens from DN patients and high-glucose-challenged podocyte models in vitro revealed a marked downregulation of HNRNP I expression relative to normal renal tissues and podocytes. In vitro assays demonstrated that high-glucose conditions precipitated a significant reduction in podocyte viability and an escalation in markers indicative of apoptosis. Conversely, HNRNP I overexpression was found to restore podocyte viability and attenuate apoptotic indices. IRAK1, a gene encoding a protein integral to inflammatory signaling, was shown to interact with HNRNP I, which promotes IRAK1 degradation. This interaction culminates in suppressing the PI3K/AKT/mTOR signaling pathway, thereby diminishing podocyte apoptosis and mitigating renal damage in DN. This investigation unveils the mechanistic role of HNRNP I in DN for the first time, potentially informing novel therapeutic strategies for DN renal impairment.


Subject(s)
Apoptosis , Diabetic Nephropathies , Interleukin-1 Receptor-Associated Kinases , Podocytes , Signal Transduction , Podocytes/metabolism , Podocytes/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Proteolysis , Inflammation , TOR Serine-Threonine Kinases/metabolism , Glucose/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/genetics
5.
Sci Rep ; 14(1): 15009, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38951638

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory bowel disease with intricate pathogenesis and varied presentation. Accurate diagnostic tools are imperative to detect and manage UC. This study sought to construct a robust diagnostic model using gene expression profiles and to identify key genes that differentiate UC patients from healthy controls. Gene expression profiles from eight cohorts, encompassing a total of 335 UC patients and 129 healthy controls, were analyzed. A total of 7530 gene sets were computed using the GSEA method. Subsequent batch correction, PCA plots, and intersection analysis identified crucial pathways and genes. Machine learning, incorporating 101 algorithm combinations, was employed to develop diagnostic models. Verification was done using four external cohorts, adding depth to the sample repertoire. Evaluation of immune cell infiltration was undertaken through single-sample GSEA. All statistical analyses were conducted using R (Version: 4.2.2), with significance set at a P value below 0.05. Employing the GSEA method, 7530 gene sets were computed. From this, 19 intersecting pathways were discerned to be consistently upregulated across all cohorts, which pertained to cell adhesion, development, metabolism, immune response, and protein regulation. This corresponded to 83 unique genes. Machine learning insights culminated in the LASSO regression model, which outperformed others with an average AUC of 0.942. This model's efficacy was further ratified across four external cohorts, with AUC values ranging from 0.694 to 0.873 and significant Kappa statistics indicating its predictive accuracy. The LASSO logistic regression model highlighted 13 genes, with LCN2, ASS1, and IRAK3 emerging as pivotal. Notably, LCN2 showcased significantly heightened expression in active UC patients compared to both non-active patients and healthy controls (P < 0.05). Investigations into the correlation between these genes and immune cell infiltration in UC highlighted activated dendritic cells, with statistically significant positive correlations noted for LCN2 and IRAK3 across multiple datasets. Through comprehensive gene expression analysis and machine learning, a potent LASSO-based diagnostic model for UC was developed. Genes such as LCN2, ASS1, and IRAK3 hold potential as both diagnostic markers and therapeutic targets, offering a promising direction for future UC research and clinical application.


Subject(s)
Colitis, Ulcerative , Machine Learning , Humans , Colitis, Ulcerative/genetics , Colitis, Ulcerative/diagnosis , Algorithms , Gene Expression Profiling/methods , Transcriptome , Interleukin-1 Receptor-Associated Kinases/genetics , Male , Female , Lipocalin-2/genetics , Case-Control Studies , Biomarkers , Adult
6.
Sci Rep ; 14(1): 16393, 2024 07 16.
Article in English | MEDLINE | ID: mdl-39014006

ABSTRACT

The search for dementia treatments, including treatments for neuropsychiatric lupus (NPSLE), has not yet uncovered useful therapeutic targets that mitigate underlying inflammation. Currently, NPSLE's limited treatment options are often accompanied by severe toxicity. Blocking toll-like receptor (TLR) and IL-1 receptor signal transduction by inhibiting interleukin-1 receptor-associated kinase 4 (IRAK4) offers a new pathway for intervention. Using a pre-clinical NPSLE model, we compare lupus-like B6.MRL-Faslpr (MRL) mice with B6.MRL-Faslpr-IRAK4 kinase-dead (MRL-IRAK4-KD) mice, which are were less prone to 'general' lupus-like symptoms. We demonstrate that lupus-prone mice with a mutation in the kinase domain of IRAK4 no longer display typical lupus hallmarks such as splenomegaly, inflammation, production of hormones, and anti-double-stranded (ds)DNA antibody. water maze behavioral testing, which measures contextual associative learning, revealed that mice without functional IRAK4 displayed a recovery in memory acquisition deficits. RNA-seq approach revealed that cytokine and hormone signaling converge on the JAK/STAT pathways in the mouse hippocampus. Ultimately, the targets identified in this work may result in broad clinical value that can fill the significant scientific and therapeutic gaps precluding development of cures for dementia.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Lupus Vasculitis, Central Nervous System , Animals , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Mice , Lupus Vasculitis, Central Nervous System/immunology , Lupus Vasculitis, Central Nervous System/metabolism , Disease Models, Animal , Female , Signal Transduction
7.
SLAS Technol ; 29(4): 100172, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39067816

ABSTRACT

Bone Marrow mesenchymal Stem Cells (BMSCs) are considered as an important source of cells for regenerative medicine, In particular, Bone Marrow mesenchymal Stem Cells Exosomes (BMSCs-EXO) have the most significant effect in the treatment of Spinal Cord Injury (SCI), but the mechanism of action is still unknown. This study found that compared with other SCI groups, BMSCs-EXO loaded with miR-146a could significantly improve the functional recovery of the hind limbs of SCI rats. Hematoxylin and eosin (H&E) indicated that the lesion area of spinal cord injury was less, nissl staining indicated that the number of nissl bodies remained more; the mechanism may be through inhibiting the expression of IRAK1 and TRAF6, blocking the activation of NF-κB p65, reducing the expression of TNF-α, IL-1ß and IL-6 inflammatory factors and oxidative stress, improving the SCI microenvironment, and promoting the repair of neural function. In general, we found that BMSCs-EXO loaded with miR-146a could reduce the inflammatory response and oxidative stress in SCI by inhibiting the activation of IRAK1/TRAF6/NF-κB p65 signaling pathway, and promote the recovery of neurological function in SCI rats.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Mesenchymal Stem Cells , MicroRNAs , Spinal Cord Injuries , TNF Receptor-Associated Factor 6 , Animals , Spinal Cord Injuries/therapy , MicroRNAs/metabolism , MicroRNAs/genetics , Mesenchymal Stem Cells/metabolism , Rats , Interleukin-1 Receptor-Associated Kinases/metabolism , TNF Receptor-Associated Factor 6/metabolism , Extracellular Vesicles/metabolism , Rats, Sprague-Dawley , Signal Transduction , Disease Models, Animal , Bone Marrow Cells
8.
Int Immunopharmacol ; 140: 112767, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39083922

ABSTRACT

The skin acts as a vital barrier, shielding the body from external threats that can trigger dryness, itching, and inflammation. Pilea mongolica, a traditional Chinese medicinal herb, holds promise for various ailments, yet its anti-inflammatory properties remain understudied. This study aimed to explore the potential anti-inflammatory effects of the methanol extract of P. mongolica (MEPM) and its underlying molecular mechanisms and active compounds in LPS-stimulated human keratinocytes. MEPM treatment, at concentrations without cytotoxicity, significantly decreased NO productions and the iNOS, IL-6, IL-1ß, and TNF-α levels in LPS-induced HaCaT cells. Moreover, MEPM suppressed IRAK4 expression and phosphorylation of JNK, ERK, p38, p65, and c-Jun, suggesting that the anti-inflammatory effects of MEPM result from the inhibition of IRAK4/MAPK/NF-κB/AP-1 signaling pathway. Through LC/MS/MS analysis, 30 compounds and 24 compounds were estimated in negative and positive modes, respectively, including various anti-inflammatory compounds, such as corilagin and geraniin. Through HPLC analysis, geraniin was found to be present in MEPM at a concentration of 18.87 mg/g. Similar to MEPM, geraniin reduced iNOS mRNA expression and inhibited NO synthesis. It also decreased mRNA and protein levels of inflammatory cytokines, including IL-6 and TNF-α, and inhibited IRAK4 expression and the phosphorylation of MAPKs, NF-κB, and AP-1 pathways. Therefore, it can be inferred that the anti-inflammatory effects of MEPM are attributable to geraniin. Thus, MEPM and its active compound geraniin are potential candidates for use in natural functional cosmetics.


Subject(s)
Anti-Inflammatory Agents , Glucosides , Hydrolyzable Tannins , Keratinocytes , Lipopolysaccharides , Plant Extracts , Signal Transduction , Humans , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Glucosides/pharmacology , HaCaT Cells , Hydrolyzable Tannins/pharmacology , Inflammation/drug therapy , Inflammation/chemically induced , Interleukin-1 Receptor-Associated Kinases/metabolism , Keratinocytes/drug effects , Keratinocytes/immunology , Methanol/chemistry , NF-kappa B/metabolism , Nitric Oxide/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism
9.
Clin Immunol ; 266: 110327, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053866

ABSTRACT

This study retrospectively investigated the impact of interleukin-1 receptor-associated kinase-3 (IRAK-3/IRAK-M) silencing by methylation on the likelihood of multiple sclerosis (MS) activity. This cross-sectional study included 90 patients with MS: 45 with active disease (Group 1), 45 in remission (Group 2), and 45 healthy controls. The study included quantitation of IRAK-3 methylation index (MI%), IRAK-3 mRNA, and myeloid differentiation factor88 (MyD88) and assessment of NF-κB activity. IRAK-3 MI% was significantly higher in group 1 compared to group 2, accompanied by lower IRAK-3 mRNA expression, elevated circulating MyD88, and increased NF-κB activity. IRAK-3 MI% correlated negatively with its transcript and positively with MyD88 and NF-κB activity. A logistic regression model was created to predict active demyelination. The C-index was 0.924, which indicates a very strong prediction model. Within the limitations of current work, IRAK-3 methylation level seems to be a promising candidate biomarker for identifying MS patients at risk of relapse.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Multiple Sclerosis , Myeloid Differentiation Factor 88 , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Female , Male , Adult , Multiple Sclerosis/genetics , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Myeloid Differentiation Factor 88/genetics , Middle Aged , Cross-Sectional Studies , NF-kappa B/metabolism , NF-kappa B/genetics , Recurrence , Retrospective Studies , DNA Methylation , Biomarkers/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
11.
Cell Death Dis ; 15(6): 390, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830885

ABSTRACT

Glioma is the most common and aggressive type of primary malignant brain tumor. The N6-methyladenosine (m6A) modification widely exists in eukaryotic cells and plays an important role in the occurrence and development of human tumors. However, the function and mechanism of heterogeneous nuclear ribonucleoprotein C (HNRNPC), an RNA-binding protein and m6A reader in gliomas remains to be comprehensively and extensively explored. Herein, we found that HNRNPC mRNA and protein overexpression were associated with a poor prognosis for patients with gliomas, based on the data from TCGA, the CGGA, and the TMAs. Biologically, HNRNPC knockdown markedly repressed malignant phenotypes of glioma in vitro and in vivo, whereas ectopic HNRNPC expression had the opposite effect. Integrative RNA sequencing and MeRIP sequencing analyses identified interleukin-1 receptor-associated kinase 1 (IRAK1) as a downstream target of HNRNPC. The glioma public datasets and tissue microarrays (TMAs) data indicated that IRAK1 overexpression was associated with poor prognosis, and IRAK1 knockdown significantly repressed malignant biological behavior in vitro. Mechanistically, HNRNPC maintains the mRNA stability of IRAK1 in an m6A-dependent manner, resulting in activation of the mitogen-activated protein kinase (MAPK) signaling pathway, which was necessary for the malignant behavior of glioma. Our findings demonstrate the HNRNPC-IRAK1-MAPK axis as a crucial carcinogenic factor for glioma and the novel underlying mechanism of IRAK1 upregulation, which provides a rationale for therapeutically targeting epitranscriptomic modulators in glioma.


Subject(s)
Disease Progression , Glioma , Heterogeneous-Nuclear Ribonucleoprotein Group C , Interleukin-1 Receptor-Associated Kinases , MAP Kinase Signaling System , RNA, Messenger , Humans , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Cell Line, Tumor , MAP Kinase Signaling System/genetics , Mice , RNA Stability/genetics , Mice, Nude , Animals , Gene Expression Regulation, Neoplastic , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Female , Male , Adenosine/analogs & derivatives , Adenosine/metabolism , Prognosis
12.
Elife ; 132024 Jun 12.
Article in English | MEDLINE | ID: mdl-38864842

ABSTRACT

The Myddosome is a key innate immune signalling platform. It forms at the cell surface and contains MyD88 and IRAK proteins which ultimately coordinate the production of pro-inflammatory cytokines. Toll-like receptor 4 (TLR4) signals via the Myddosome when triggered by lipopolysaccharide (LPS) or amyloid-beta (Aß) aggregates but the magnitude and time duration of the response are very different for reasons that are unclear. Here, we followed the formation of Myddosomes in live macrophages using local delivery of TLR4 agonist to the cell surface and visualisation with 3D rapid light sheet imaging. This was complemented by super-resolution imaging of Myddosomes in fixed macrophages to determine the size of the signalling complex at different times after triggering. Myddosomes formed more rapidly after LPS than in response to sonicated Aß 1-42 fibrils (80 vs 372 s). The mean lifetimes of the Myddosomes were also shorter when triggered by LPS compared to sonicated Aß fibrils (170 and 220 s), respectively. In both cases, a range of Myddosome of different sizes (50-500 nm) were formed. In particular, small round Myddosomes around 100 nm in size formed at early time points, then reduced in proportion over time. Collectively, our data suggest that compared to LPS the multivalency of Aß fibrils leads to the formation of larger Myddosomes which form more slowly and, due to their size, take longer to disassemble. This explains why sonicated Aß fibrils results in less efficient triggering of TLR4 signalling and may be a general property of protein aggregates.


Subject(s)
Amyloid beta-Peptides , Lipopolysaccharides , Myeloid Differentiation Factor 88 , Toll-Like Receptor 4 , Toll-Like Receptor 4/metabolism , Amyloid beta-Peptides/metabolism , Myeloid Differentiation Factor 88/metabolism , Animals , Mice , Kinetics , Macrophages/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Signal Transduction
13.
Sci Transl Med ; 16(750): eadi4125, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38838135

ABSTRACT

Chronic inflammation is a constitutive component of many age-related diseases, including age-related macular degeneration (AMD). Here, we identified interleukin-1 receptor-associated kinase M (IRAK-M) as a key immunoregulator in retinal pigment epithelium (RPE) that declines during the aging process. Rare genetic variants of IRAK3, which encodes IRAK-M, were associated with an increased likelihood of developing AMD. In human samples and mouse models, IRAK-M abundance in the RPE declined with advancing age or exposure to oxidative stress and was further reduced in AMD. Irak3-knockout mice exhibited an increased incidence of outer retinal degeneration at earlier ages, which was further exacerbated by oxidative stressors. The absence of IRAK-M led to a disruption in RPE cell homeostasis, characterized by compromised mitochondrial function, cellular senescence, and aberrant cytokine production. IRAK-M overexpression protected RPE cells against oxidative or immune stressors. Subretinal delivery of adeno-associated virus (AAV)-expressing human IRAK3 rescued light-induced outer retinal degeneration in wild-type mice and attenuated age-related spontaneous retinal degeneration in Irak3-knockout mice. Our data show that replenishment of IRAK-M in the RPE may redress dysregulated pro-inflammatory processes in AMD, suggesting a potential treatment for retinal degeneration.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Mice, Knockout , Oxidative Stress , Retinal Degeneration , Retinal Pigment Epithelium , Animals , Humans , Male , Mice , Cellular Senescence , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
14.
J Immunol ; 213(3): 362-372, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38847613

ABSTRACT

IL-1R-associated kinases (IRAKs) are signal transducers of the TLR/IL-1R-MyD88-TRAF6 pathways. Vertebrates possess two IRAK lineages, IRAK1/2/3 and IRAK4. In mammals, IRAK4/IRAK1 and IRAK4/IRAK2 are pathway enhancers, whereas IRAK3 is a repressor. However, in bony fish, IRAK2 is absent, and it remains elusive how fish IRAK1/3/4 functionally differ from their mammalian counterparts. In this study, we explored this using the zebrafish model. First, we showed that in human 293T cells, zebrafish IRAK1 and IRAK4 were components of the Myddosome (MyD88-IRAK4-IRAK1) complex, with IRAK1 serving as a potent pathway enhancer. Then, we discovered two zebrafish IRAK3 variants: one (IRAK3a) contains an N-terminal Death domain, a middle pseudokinase domain, and a C-terminal TRAF6-binding domain, whereas the other (IRAK3b) lost both the kinase and TRAF6-binding domains. This truncation of IRAK3 variants could be a conserved phenomenon in fish, because it is also observed in trout and grass carp. We proceeded to show that zebrafish IRAK3a acts as a pathway enhancer by binding with MyD88 and TRAF6, but its activity is milder than IRAK1, possibly because it has no kinase activity. Zebrafish IRAK3b, however, plays a sheer negative role, apparently because of its lack of kinase and TRAF6-binding domains. Moreover, zebrafish IRAK3a/3b inhibit the activity of IRAK1/4, not by interacting with IRAK1/4 but possibly by competing for MyD88 and TRAF6. Finally, we have verified the essential activities of zebrafish IRAK1/3a/3b/4 in zebrafish cells and embryos. In summary, to our knowledge, our findings provide new insights into the molecular functions of fish IRAKs and the evolution of the IRAK functional modes in vertebrates.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Myeloid Differentiation Factor 88 , Signal Transduction , TNF Receptor-Associated Factor 6 , Zebrafish Proteins , Zebrafish , Animals , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Humans , Signal Transduction/immunology , HEK293 Cells , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
15.
J Med Chem ; 67(13): 10687-10709, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38913701

ABSTRACT

UC and ALI are inflammatory diseases with limited treatment in the clinic. Herein, fragment-based anti-inflammatory agent designs were carried out deriving from cyclohexylamine/cyclobutylamine and several fragments from anti-inflammatory agents in our lab. AF-45 (IC50 = 0.53/0.60 µM on IL-6/TNF-α in THP-1 macrophages) was identified as the optimal molecule using ELISA and MTT assays from the 33 synthesized compounds. Through mechanistic studies and a systematic target search process, AF-45 was found to block the NF-κB/MAPK pathway and target IRAK4, a promising target for inflammation and autoimmune diseases. The selectivity of AF-45 targeting IRAK4 was validated by comparing its effects on other kinase/nonkinase proteins. In vivo, AF-45 exhibited a good therapeutic effect on UC and ALI, and favorable PK proprieties. Since there are currently no clinical or preclinical trials for IRAK4 inhibitors to treat UC and ALI, AF-45 provides a new lead compound or candidate targeting IRAK4 for the treatment of these diseases.


Subject(s)
Acute Lung Injury , Colitis, Ulcerative , Interleukin-1 Receptor-Associated Kinases , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Humans , Animals , Colitis, Ulcerative/drug therapy , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Drug Design , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Drug Discovery , Male , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Structure-Activity Relationship , THP-1 Cells
16.
J Med Chem ; 67(13): 10548-10566, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38920289

ABSTRACT

Developing therapies for the activated B-cell like (ABC) subtype of diffuse large B-cell lymphomas (DLBCL) remains an area of unmet medical need. A subset of ABC DLBCL tumors is driven by activating mutations in myeloid differentiation primary response protein 88 (MYD88), which lead to constitutive activation of interleukin-1 receptor associated kinase 4 (IRAK4) and cellular proliferation. IRAK4 signaling is driven by its catalytic and scaffolding functions, necessitating complete removal of this protein and its escape mechanisms for complete therapeutic suppression. Herein, we describe the identification and characterization of a dual-functioning molecule, KT-413 and show it efficiently degrades IRAK4 and the transcription factors Ikaros and Aiolos. KT-413 achieves concurrent degradation of these proteins by functioning as both a heterobifunctional degrader and a molecular glue. Based on the demonstrated activity and safety of KT-413 in preclinical studies, a phase 1 clinical trial in B-cell lymphomas, including MYD88 mutant ABC DLBCL, is currently underway.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Lymphoma, Large B-Cell, Diffuse , Mutation , Myeloid Differentiation Factor 88 , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Humans , Animals , Cell Line, Tumor , Drug Discovery , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Mice , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazoles/metabolism , Proteolysis/drug effects , Structure-Activity Relationship
17.
Clin Immunol ; 265: 110268, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838930

ABSTRACT

PURPOSE: To report a case of a five-month-old Chinese infant who died of interleukin-1 receptor-associated kinase-4 (IRAK-4) deficiency presenting with rapid and progressive Pseudomonas aeruginosa sepsis. METHODS: The genetic etiology of IRAK-4 deficiency was confirmed through trio-whole exome sequencing and Sanger sequencing. Functional consequences were invested using an in vitro minigene splicing assay. RESULTS: Trio-whole exome sequencing of genomic DNA identified two novel compound heterozygous mutations, IRAK-4 (NM_016123.3): c.942-1G > A and c.644_651+ 6delTTGCAGCAGTAAGT in the proband, which originated from his symptom-free parents. These mutations were predicted to cause frameshifts and generate three truncated proteins without enzyme activity. CONCLUSIONS: Our findings expand the range of IRAK-4 mutations and provide functional support for the pathogenic effects of splice-site mutations. Additionally, this case highlights the importance of considering the underlying genetic defects of immunity when dealing with unusually overwhelming infections in previously healthy children and emphasizes the necessity for timely treatment with wide-spectrum antimicrobials.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Pseudomonas Infections , Pseudomonas aeruginosa , Sepsis , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/deficiency , Pseudomonas aeruginosa/genetics , Pseudomonas Infections/genetics , Male , Infant , Sepsis/genetics , Sepsis/microbiology , Primary Immunodeficiency Diseases/genetics , Loss of Function Mutation , Heterozygote , Exome Sequencing , Immunologic Deficiency Syndromes/genetics
18.
Eur J Pharmacol ; 978: 176773, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38936453

ABSTRACT

The interleukin-1 receptor-associated kinase (IRAK) family is a group of serine-threonine kinases that regulates various cellular processes via toll-like receptor (TLR)/interleukin-1 receptor (IL1R)-mediated signaling. The IRAK family comprises four members, including IRAK1, IRAK2, IRAK3, and IRAK4, which play an important role in the expression of various inflammatory genes, thereby contributing to the inflammatory response. IRAKs are key proteins in chronic and acute liver diseases, and recent evidence has implicated IRAK family proteins (IRAK1, IRAK3, and IRAK4) in the progression of liver-related disorders, including alcoholic liver disease, non-alcoholic steatohepatitis, hepatitis virus infection, acute liver failure, liver ischemia-reperfusion injury, and hepatocellular carcinoma. In this article, we provide a comprehensive review of the role of IRAK family proteins and their associated inflammatory signaling pathways in the pathogenesis of liver diseases. The purpose of this study is to explore whether IRAK family proteins can serve as the main target for the treatment of liver related diseases.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Liver Diseases , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Humans , Liver Diseases/metabolism , Animals , Signal Transduction
19.
Dis Model Mech ; 17(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38881329

ABSTRACT

MECP2 duplication syndrome (MDS) is a neurodevelopmental disorder caused by tandem duplication of the MECP2 locus and its surrounding genes, including IRAK1. Current MDS mouse models involve transgenic expression of MECP2 only, limiting their applicability to the study of the disease. Herein, we show that an efficient and precise CRISPR/Cas9 fusion proximity-based approach can be utilized to generate an Irak1-Mecp2 tandem duplication mouse model ('Mecp2 Dup'). The Mecp2 Dup mouse model recapitulates the genomic landscape of human MDS by harboring a 160 kb tandem duplication encompassing Mecp2 and Irak1, representing the minimal disease-causing duplication, and the neighboring genes Opn1mw and Tex28. The Mecp2 Dup model exhibits neuro-behavioral abnormalities, and an abnormal immune response to infection not previously observed in other mouse models, possibly owing to Irak1 overexpression. The Mecp2 Dup model thus provides a tool to investigate MDS disease mechanisms and develop potential therapies applicable to patients.


Subject(s)
Disease Models, Animal , Gene Duplication , Interleukin-1 Receptor-Associated Kinases , Mental Retardation, X-Linked , Methyl-CpG-Binding Protein 2 , Animals , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Humans , Mice, Inbred C57BL , Mice , CRISPR-Cas Systems/genetics , Behavior, Animal , Male
20.
Eur J Med Chem ; 272: 116487, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38759452

ABSTRACT

Acute lung injury (ALI) and inflammatory bowel disease (IBD) are common inflammatory illnesses that seriously affect people's health. Herein, a series of 4-hydroxylcoumarin (4-HC) derivatives were designed and synthesized. The inhibitory effects of these compounds on LPS-induced interleukin-6 (IL-6) release from J774A.1 cells were then screened via ELISA assay, compound B8 showed 3 times more active than the lead compound 4-HC. The most active compound B8 had the IC50 values of 4.57 µM and 6.51 µM for IL-6 release on mouse cells J774A.1 and human cells THP-1, respectively. Furthermore, we also found that B8 could act on the MAPK pathway. Based on the target prediction results of computer virtual docking, kinase inhibitory assay was carried out, and it revealed that targeting IRAK1 was a key mechanism for B8 to exert anti-inflammatory activity. Moreover, B8 exerted a good therapeutic effect on the dextran sulfate sodium (DSS)-induced colitis model and liposaccharide (LPS)-induced ALI mouse models. The acute toxicity experiments indicated that high-dose B8 caused no adverse reactions in mice, confirming its safety in vivo. Additionally, the preliminary pharmacokinetic (PK) parameters of B8 in SD rats were also examined, revealing a bioavailability (F) of 28.72 %. In conclusion, B8 is a potential candidate of drug for the treatment of ALI and colitis.


Subject(s)
4-Hydroxycoumarins , Acute Lung Injury , Colitis , Drug Design , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Animals , Colitis/drug therapy , Colitis/chemically induced , Mice , Humans , Structure-Activity Relationship , 4-Hydroxycoumarins/pharmacology , 4-Hydroxycoumarins/chemistry , 4-Hydroxycoumarins/chemical synthesis , Molecular Structure , Dextran Sulfate , Male , Dose-Response Relationship, Drug , Rats , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Interleukin-6/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Molecular Docking Simulation , Mice, Inbred C57BL , Cell Line
SELECTION OF CITATIONS
SEARCH DETAIL