Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.701
Filtrer
1.
Sci Rep ; 14(1): 16393, 2024 07 16.
Article de Anglais | MEDLINE | ID: mdl-39014006

RÉSUMÉ

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.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Vascularite lupique du système nerveux central , Animaux , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , Souris , Vascularite lupique du système nerveux central/immunologie , Vascularite lupique du système nerveux central/métabolisme , Modèles animaux de maladie humaine , Femelle , Transduction du signal
2.
Sci Rep ; 14(1): 15009, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38951638

RÉSUMÉ

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.


Sujet(s)
Rectocolite hémorragique , Apprentissage machine , Humains , Rectocolite hémorragique/génétique , Rectocolite hémorragique/diagnostic , Algorithmes , Analyse de profil d'expression de gènes/méthodes , Transcriptome , Interleukin-1 Receptor-Associated Kinases/génétique , Mâle , Femelle , Lipocaline-2/génétique , Études cas-témoins , Marqueurs biologiques , Adulte
3.
Dis Model Mech ; 17(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38881329

RÉSUMÉ

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.


Sujet(s)
Modèles animaux de maladie humaine , Duplication de gène , Interleukin-1 Receptor-Associated Kinases , Retard mental lié à l'X , Protéine-2 de liaison au CpG méthylé , Animaux , Protéine-2 de liaison au CpG méthylé/génétique , Protéine-2 de liaison au CpG méthylé/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , Interleukin-1 Receptor-Associated Kinases/métabolisme , Retard mental lié à l'X/génétique , Retard mental lié à l'X/anatomopathologie , Humains , Souris de lignée C57BL , Souris , Systèmes CRISPR-Cas/génétique , Comportement animal , Mâle
4.
Eur J Pharmacol ; 978: 176773, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38936453

RÉSUMÉ

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.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Maladies du foie , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , Humains , Maladies du foie/métabolisme , Animaux , Transduction du signal
5.
J Med Chem ; 67(13): 10687-10709, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38913701

RÉSUMÉ

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.


Sujet(s)
Lésion pulmonaire aigüe , Rectocolite hémorragique , Interleukin-1 Receptor-Associated Kinases , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/métabolisme , Humains , Animaux , Rectocolite hémorragique/traitement médicamenteux , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/usage thérapeutique , Conception de médicament , Souris , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/synthèse chimique , Anti-inflammatoires/composition chimique , Découverte de médicament , Mâle , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Relation structure-activité , Cellules THP-1
6.
J Med Chem ; 67(13): 10548-10566, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38920289

RÉSUMÉ

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.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Lymphome B diffus à grandes cellules , Mutation , Facteur de différenciation myéloïde-88 , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Facteur de différenciation myéloïde-88/métabolisme , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/métabolisme , Lymphome B diffus à grandes cellules/anatomopathologie , Humains , Animaux , Lignée cellulaire tumorale , Découverte de médicament , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Souris , Imidazoles/composition chimique , Imidazoles/pharmacologie , Imidazoles/métabolisme , Protéolyse/effets des médicaments et des substances chimiques , Relation structure-activité
7.
J Immunol ; 213(3): 362-372, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38847613

RÉSUMÉ

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.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Facteur de différenciation myéloïde-88 , Transduction du signal , Facteur-6 associé aux récepteurs de TNF , Protéines de poisson-zèbre , Danio zébré , Animaux , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Facteur-6 associé aux récepteurs de TNF/métabolisme , Facteur-6 associé aux récepteurs de TNF/génétique , Humains , Transduction du signal/immunologie , Cellules HEK293 , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique
8.
Clin Immunol ; 265: 110268, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38838930

RÉSUMÉ

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.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Infections à Pseudomonas , Pseudomonas aeruginosa , Sepsie , Humains , Interleukin-1 Receptor-Associated Kinases/génétique , Interleukin-1 Receptor-Associated Kinases/déficit , Pseudomonas aeruginosa/génétique , Infections à Pseudomonas/génétique , Mâle , Nourrisson , Sepsie/génétique , Sepsie/microbiologie , Maladies d'immunodéficience primaire/génétique , Mutation perte de fonction , Hétérozygote , , Déficits immunitaires/génétique
9.
Elife ; 132024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38864842

RÉSUMÉ

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.


Sujet(s)
Peptides bêta-amyloïdes , Lipopolysaccharides , Facteur de différenciation myéloïde-88 , Récepteur de type Toll-4 , Récepteur de type Toll-4/métabolisme , Peptides bêta-amyloïdes/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Animaux , Souris , Cinétique , Macrophages/métabolisme , Interleukin-1 Receptor-Associated Kinases/métabolisme , Transduction du signal
10.
Cell Death Dis ; 15(6): 390, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830885

RÉSUMÉ

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.


Sujet(s)
Évolution de la maladie , Gliome , Ribonucléoprotéine nucléaire hétérogène du groupe C , Interleukin-1 Receptor-Associated Kinases , Système de signalisation des MAP kinases , ARN messager , Humains , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , ARN messager/métabolisme , ARN messager/génétique , Ribonucléoprotéine nucléaire hétérogène du groupe C/métabolisme , Ribonucléoprotéine nucléaire hétérogène du groupe C/génétique , Lignée cellulaire tumorale , Système de signalisation des MAP kinases/génétique , Souris , Stabilité de l'ARN/génétique , Souris nude , Animaux , Régulation de l'expression des gènes tumoraux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Femelle , Mâle , Adénosine/analogues et dérivés , Adénosine/métabolisme , Pronostic
11.
Sci Transl Med ; 16(750): eadi4125, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38838135

RÉSUMÉ

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.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Souris knockout , Stress oxydatif , Dégénérescence de la rétine , Épithélium pigmentaire de la rétine , Animaux , Humains , Mâle , Souris , Vieillissement de la cellule , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/génétique , Dégénérescence maculaire/métabolisme , Dégénérescence maculaire/anatomopathologie , Dégénérescence maculaire/génétique , Souris de lignée C57BL , Mitochondries/métabolisme , Dégénérescence de la rétine/métabolisme , Dégénérescence de la rétine/anatomopathologie , Dégénérescence de la rétine/génétique , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie
13.
Cell Death Dis ; 15(5): 362, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38796478

RÉSUMÉ

Advanced epithelial ovarian cancer (EOC) survival rates are dishearteningly low, with ~25% surviving beyond 5 years. Evidence suggests that cancer stem cells contribute to acquired chemoresistance and tumor recurrence. Here, we show that IRAK1 is upregulated in EOC tissues, and enhanced expression correlates with poorer overall survival. Moreover, low molecular weight hyaluronic acid, which is abundant in malignant ascites from patients with advanced EOC, induced IRAK1 phosphorylation leading to STAT3 activation and enhanced spheroid formation. Knockdown of IRAK1 impaired tumor growth in peritoneal disease models, and impaired HA-induced spheroid growth and STAT3 phosphorylation. Finally, we determined that TCS2210, a known inducer of neuronal differentiation in mesenchymal stem cells, is a selective inhibitor of IRAK1. TCS2210 significantly inhibited EOC growth in vitro and in vivo both as monotherapy, and in combination with cisplatin. Collectively, these data demonstrate IRAK1 as a druggable target for EOC.


Sujet(s)
Carcinome épithélial de l'ovaire , Acide hyaluronique , Interleukin-1 Receptor-Associated Kinases , Cellules souches tumorales , Tumeurs de l'ovaire , Facteur de transcription STAT-3 , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Humains , Facteur de transcription STAT-3/métabolisme , Femelle , Carcinome épithélial de l'ovaire/métabolisme , Carcinome épithélial de l'ovaire/anatomopathologie , Carcinome épithélial de l'ovaire/traitement médicamenteux , Carcinome épithélial de l'ovaire/génétique , Acide hyaluronique/métabolisme , Acide hyaluronique/pharmacologie , Animaux , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/génétique , Cellules souches tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Lignée cellulaire tumorale , Souris , Cisplatine/pharmacologie , Souris nude , Phosphorylation/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Masse moléculaire , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Molecules ; 29(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38792088

RÉSUMÉ

Interleukin receptor-associated kinase (IRAK) proteins are pivotal in interleukin-1 and Toll-like receptor-mediated signaling pathways. They play essential roles in innate immunity and inflammation. This review analyzes and discusses the physiological functions of IRAK1 and its associated diseases. IRAK1 is involved in a wide range of diseases such as dry eye, which highlights its potential as a therapeutic target under various conditions. Various IRAK1 inhibitors, including Pacritinib and Rosoxacin, show therapeutic potential against malignancies and inflammatory diseases. The covalent IRAK1 inhibitor JH-X-119-01 shows promise in B-cell lymphomas, emphasizing the significance of covalent bonds in its activity. Additionally, the emergence of selective IRAK1 degraders, such as JNJ-101, provides a novel strategy by targeting the scaffolding function of IRAK1. Thus, the evolving landscape of IRAK1-targeted approaches provides promising avenues for increasingly safe and effective therapeutic interventions for various diseases.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Interleukin-1 Receptor-Associated Kinases/métabolisme , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Humains , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Animaux , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme
15.
Eur J Med Chem ; 272: 116487, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38759452

RÉSUMÉ

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.


Sujet(s)
4-Hydroxycoumarines , Lésion pulmonaire aigüe , Colite , Conception de médicament , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/induit chimiquement , Animaux , Colite/traitement médicamenteux , Colite/induit chimiquement , Souris , Humains , Relation structure-activité , 4-Hydroxycoumarines/pharmacologie , 4-Hydroxycoumarines/composition chimique , 4-Hydroxycoumarines/synthèse chimique , Structure moléculaire , Sulfate dextran , Mâle , Relation dose-effet des médicaments , Rats , Lipopolysaccharides/pharmacologie , Lipopolysaccharides/antagonistes et inhibiteurs , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/synthèse chimique , Anti-inflammatoires/composition chimique , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires non stéroïdiens/pharmacologie , Anti-inflammatoires non stéroïdiens/synthèse chimique , Anti-inflammatoires non stéroïdiens/composition chimique , Interleukine-6/métabolisme , Interleukine-6/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/métabolisme , Simulation de docking moléculaire , Souris de lignée C57BL , Lignée cellulaire
16.
J Med Chem ; 67(10): 8383-8395, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38695469

RÉSUMÉ

Interleukin receptor associated kinase 4 (IRAK4) plays an important role in innate immune signaling through Toll-like and interleukin-1 receptors and represents an attractive target for the treatment of inflammatory diseases and cancer. We previously reported the development of a potent, selective, and brain-penetrant imidazopyrimidine series of IRAK4 inhibitors. However, lead molecule BIO-7488 (1) suffered from low solubility which led to variable PK, compound accumulation, and poor in vivo tolerability. Herein, we describe the discovery of a series of pyridone analogs with improved solubility which are highly potent, selective and demonstrate desirable PK profiles including good oral bioavailability and excellent brain penetration. BIO-8169 (2) reduced the in vivo production of pro-inflammatory cytokines, was well tolerated in safety studies in rodents and dog at margins well above the predicted efficacious exposure and showed promising results in a mouse model for multiple sclerosis.


Sujet(s)
Encéphale , Interleukin-1 Receptor-Associated Kinases , Inhibiteurs de protéines kinases , Animaux , Chiens , Mâle , Souris , Rats , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Découverte de médicament , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Pyrimidines/pharmacologie , Pyrimidines/composition chimique , Pyrimidines/pharmacocinétique , Pyrimidines/synthèse chimique , Pyrimidines/usage thérapeutique , Relation structure-activité
17.
J Med Chem ; 67(10): 8060-8076, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38722184

RÉSUMÉ

Interleukin-1 receptor-associated kinase 4 (IRAK4) is a promising therapeutic target in inflammation-related diseases. However, the inhibition of IRAK4 kinase activity may lead to moderate anti-inflammatory efficacy owing to the dual role of IRAK4 as an active kinase and a scaffolding protein. Herein, we report the design, synthesis, and biological evaluation of an efficient and selective IRAK4 proteolysis-targeting chimeric molecule that eliminates IRAK4 scaffolding functions. The most potent compound, LC-MI-3, effectively degraded cellular IRAK4, with a half-maximal degradation concentration of 47.3 nM. LC-MI-3 effectively inhibited the activation of downstream nuclear factor-κB signaling and exerted more potent pharmacological effects than traditional kinase inhibitors. Furthermore, LC-MI-3 exerted significant therapeutic effects in lipopolysaccharide- and Escherichia coli-induced acute and chronic inflammatory skin models compared with kinase inhibitors in vivo. Therefore, LC-MI-3 is a candidate IRAK4 degrader in alternative targeting strategies and advanced drug development.


Sujet(s)
Interleukin-1 Receptor-Associated Kinases , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/métabolisme , Animaux , Humains , Souris , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/usage thérapeutique , Administration par voie orale , Lipopolysaccharides/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/composition chimique , Anti-inflammatoires/synthèse chimique , Anti-inflammatoires/pharmacocinétique , Biodisponibilité , Découverte de médicament , Protéolyse/effets des médicaments et des substances chimiques , Relation structure-activité , Mâle , Souris de lignée C57BL
19.
Molecules ; 29(8)2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38675622

RÉSUMÉ

IRAK4 is a critical mediator in NF-κB-regulated inflammatory signaling and has emerged as a promising therapeutic target for the treatment of autoimmune diseases; however, none of its inhibitors have received FDA approval. In this study, we identified a novel small-molecule IRAK4 kinase inhibitor, DW18134, with an IC50 value of 11.2 nM. DW18134 dose-dependently inhibited the phosphorylation of IRAK4 and IKK in primary peritoneal macrophages and RAW264.7 cells, inhibiting the secretion of TNF-α and IL-6 in both cell lines. The in vivo study demonstrated the efficacy of DW18134, significantly attenuating behavioral scores in an LPS-induced peritonitis model. Mechanistically, DW18134 reduced serum TNF-α and IL-6 levels and attenuated inflammatory tissue injury. By directly blocking IRAK4 activation, DW18134 diminished liver macrophage infiltration and the expression of related inflammatory cytokines in peritonitis mice. Additionally, in the DSS-induced colitis model, DW18134 significantly reduced the disease activity index (DAI) and normalized food and water intake and body weight. Furthermore, DW18134 restored intestinal damage and reduced inflammatory cytokine expression in mice by blocking the IRAK4 signaling pathway. Notably, DW18134 protected DSS-threatened intestinal barrier function by upregulating tight junction gene expression. In conclusion, our findings reported a novel IRAK4 inhibitor, DW18134, as a promising candidate for treating inflammatory diseases, including peritonitis and IBD.


Sujet(s)
Maladies inflammatoires intestinales , Interleukin-1 Receptor-Associated Kinases , Péritonite , Animaux , Interleukin-1 Receptor-Associated Kinases/antagonistes et inhibiteurs , Interleukin-1 Receptor-Associated Kinases/métabolisme , Souris , Péritonite/traitement médicamenteux , Péritonite/induit chimiquement , Cellules RAW 264.7 , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Macrophages péritonéaux/effets des médicaments et des substances chimiques , Macrophages péritonéaux/métabolisme , Humains , Mâle , Phosphorylation/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée C57BL
20.
Cell Signal ; 119: 111192, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38685522

RÉSUMÉ

IRAK1 has been implicated in promoting development of various types of cancers and mediating radioresistance. However, its role in cervical cancer tumorigenesis and radioresistance, as well as the potential underlying mechanisms, remain poorly defined. In this study, we evaluated IRAK1 expression in radiotherapy-treated cervical cancer tissues and found that IRAK1 expression is negatively associated with the efficacy of radiotherapy. Consistently, ionizing radiation (IR)-treated HeLa and SiHa cervical cancer cells express a lower level of IRAK1 than control cells. Depletion of IRAK1 resulted in reduced activation of the NF-κB pathway, decreased cell viability, downregulated colony formation efficiency, cell cycle arrest, increased apoptosis, and impaired migration and invasion in IR-treated cervical cancer cells. Conversely, overexpressing IRAK1 mitigated the anti-cancer effects of IR in cervical cancer cells. Notably, treatment of IRAK1-overexpressing IR-treated HeLa and SiHa cells with the NF-κB pathway inhibitor pyrrolidine dithiocarbamate (PDTC) partially counteracted the effects of excessive IRAK1. Furthermore, our study demonstrated that IRAK1 deficiency enhanced the anti-proliferative role of IR treatment in a xenograft mouse model. These collective observations highlight IRAK1's role in mitigating the anti-cancer effects of radiotherapy, partly through the activation of the NF-κB pathway. SUMMARY: IRAK1 enhances cervical cancer resistance to radiotherapy, with IR treatment reducing IRAK1 expression and increasing cancer cell vulnerability and apoptosis.


Sujet(s)
Apoptose , Interleukin-1 Receptor-Associated Kinases , Facteur de transcription NF-kappa B , Tumeurs du col de l'utérus , Interleukin-1 Receptor-Associated Kinases/métabolisme , Humains , Tumeurs du col de l'utérus/radiothérapie , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme , Femelle , Animaux , Facteur de transcription NF-kappa B/métabolisme , Apoptose/effets des radiations , Souris , Cellules HeLa , Prolifération cellulaire , Souris nude , Lignée cellulaire tumorale , Transduction du signal , Mouvement cellulaire , Radiotolérance , Tests d'activité antitumorale sur modèle de xénogreffe , Survie cellulaire/effets des radiations , Rayonnement ionisant
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE