Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Arch Dermatol Res ; 316(7): 478, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023568

ABSTRACT

The efficacy of ritlecitinib, an oral JAK3/TEC family kinase inhibitor, on active and stable lesions was evaluated in patients with active non-segmental vitiligo in a phase 2b trial (NCT03715829). Patients were randomized to placebo or daily ritlecitinib 50 mg (with or without 4-week 100-mg or 200-mg loading dose), 30 mg, or 10 mg for 24 weeks. Active lesions showed greater baseline expression of inflammatory/immune markers IFNG and CCL5, levels of CD103, and T-cell infiltrates than stable lesions. Patients with more active than stable vitiligo lesions showed higher baseline serum levels of CXCL9 and PD-L1, while patients with more stable than active lesions showed higher baseline serum levels of HO-1. At Week 24, ritlecitinib 50 mg significantly stabilized mean percent change from baseline in depigmentation extent in both active lesions and stable lesions vs. placebo-response, with stable lesions showing greater repigmentation. After 24 weeks of treatment, ritlecitinib 50 mg increased expression of melanocyte markers in stable lesions, while Th1/Th2-related and co-stimulatory molecules decreased significantly in both stable and active lesions. Serum from patients with more active than stable lesions showed decreased levels of ICOS and NK cell activation markers. These data, confirmed at transcription/protein levels, indicate that stable lesion repigmentation occurs early with ritlecitinib, while active lesions require stabilization of inflammation first. ClinicalTrials.gov: NCT03715829.


Subject(s)
Janus Kinase 3 , Protein Kinase Inhibitors , Vitiligo , Humans , Vitiligo/drug therapy , Vitiligo/diagnosis , Vitiligo/immunology , Male , Female , Adult , Janus Kinase 3/antagonists & inhibitors , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/administration & dosage , Treatment Outcome , Chemokine CXCL9/blood , Chemokine CCL5/blood , Young Adult , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , B7-H1 Antigen/blood , Melanocytes/drug effects , Double-Blind Method , Skin Pigmentation/drug effects , Administration, Oral , Interferon-gamma
2.
Molecules ; 29(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38893334

ABSTRACT

Thiazolin-4-ones and their derivatives represent important heterocyclic scaffolds with various applications in medicinal chemistry. For that reason, the synthesis of two 5-substituted thiazolidin-4-one derivatives was performed. Their structure assignment was conducted by NMR experiments (2D-COSY, 2D-NOESY, 2D-HSQC and 2D-HMBC) and conformational analysis was conducted through Density Functional Theory calculations and 2D-NOESY. Conformational analysis showed that these two molecules adopt exo conformation. Their global minimum structures have two double bonds (C=N, C=C) in Z conformation and the third double (C=N) in E. Our DFT results are in agreement with the 2D-NMR measurements. Furthermore, the reaction isomerization paths were studied via DFT to check the stability of the conformers. Finally, some potential targets were found through the SwissADME platform and docking experiments were performed. Both compounds bind strongly to five macromolecules (triazoloquinazolines, mglur3, Jak3, Danio rerio HDAC6 CD2, acetylcholinesterase) and via SwissADME it was found that these two molecules obey Lipinski's Rule of Five.


Subject(s)
Molecular Conformation , Molecular Docking Simulation , Thiazolidines , Thiazolidines/chemistry , Thiazolidines/chemical synthesis , Isomerism , Animals , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Zebrafish , Magnetic Resonance Spectroscopy , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Janus Kinase 3/chemistry , Molecular Structure
3.
Transpl Immunol ; 85: 102075, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38936745

ABSTRACT

BACKGROUND: Despite the significant role of JAK3 in various autoimmune diseases, including graft-versus-host disease (GVHD), there has been a lack of potent and selective JAK3 inhibitors specifically studied for GVHD. In our preclinical investigations, we evaluated a novel JAK3 inhibitor called CS12192, which is already undergoing clinical investigation in autoimmune diseases. METHODS: We evaluated the efficacy of CS12192 in GVHD through mixed lymphocyte reaction (MLR) in both mouse and human cells, as well as allogeneic bone marrow transplantation (BMT) in a murine model. RESULTS: CS12192, starting at a concentration of 0.5 µM, dose-dependently reduced the intracellular positivity for cytokines TNF-α and IFN-γ in CD4+ T cells (p < 0.05 to p < 0.0001) and CD8+ T cells (p < 0.01 to p < 0.0001) during mouse allogeneic MLR assays. This effect was observed for both single and double positivity of the cytokines. Moreover, In MLR assays with three different human donors, CS12192 also demonstrated a dose-dependent reduction in the proportion of IFN-γ positive CD4+ T cells (p < 0.0001) and CD8+ T cells (p < 0.01 to p < 0.0001). Additionally, it suppressed T cell proliferation in the mouse MLR (p < 0.05 to p < 0.0001), but this effect was observed in only one human donor (p < 0.001 to p < 0.0001). Furthermore, the administration of CS12192 at 40 and 80 mg/kg BID significantly improved the survival rate in the BMT model, resulting in cumulative 62-day survival rates of 88.89% (p < 0.01) and 100% (p < 0.001), respectively, compared with prednisolone (p < 0.05). CONCLUSIONS: CS12192 is a novel, potent and selective JAK3 inhibitor demonstrating great potential to mitigate acute GVHD.


Subject(s)
Bone Marrow Transplantation , Graft vs Host Disease , Janus Kinase 3 , Animals , Humans , Mice , Acute Disease , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Disease Models, Animal , Graft vs Host Disease/drug therapy , Interferon-gamma/metabolism , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Lymphocyte Culture Test, Mixed , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitriles/therapeutic use , Pyrimidines/therapeutic use , Pyrimidines/pharmacology , Transplantation, Homologous , Tumor Necrosis Factor-alpha/metabolism
4.
Immunopharmacol Immunotoxicol ; 46(4): 529-537, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38918174

ABSTRACT

OBJECTIVE: Autoimmune dermatosis (AID) occurs when the body's immune system attacks skin or tissue, leading to various types of skin disorders or injuries. Recent studies show that Janus kinases (JAKs) play critical roles in autoimmune diseases including AID by regulating multiple cytokine signaling pathways. CS12192, a novel JAK3/JAK1/TBK1 inhibitor, has been reported to exert ameliorative effects in rheumatoid arthritis. However, the efficacy of CS12192 on AID is undetermined. This study aims to investigate the therapeutic efficacy of CS12192 on psoriasis (PSO), systemic lupus erythematosus (SLE) and atopic dermatitis (AD) in mouse models. METHODS: Interleukin-23 (IL-23)-induced PSO model, spontaneous SLE model of MRL/MpJ-Faslpr/J (MRL/lpr) mice, and oxazolone (OXA) and dinitrochlorobenzene (DNCB)-induced murine AD models were used for the evaluation of curative effects of CS12192, respectively. The skin lesion, biochemical parameters, ear thickness, ear weight and histopathology were assessed accordingly. RESULTS: In PSO model, mice treated with CS12192 show reduced ear thickness and ear weight as compared with vehicle. In SLE model, CS12192 ameliorates cutaneous parameters such as lymphadenectasis and skin lesion but not systematic parameters such as proteinuria concentration and score, serum dsDNA and BUN concentration. In AD models, CS12192 dose-dependently improves ear swelling and reduces histological scores, exerting equivalent efficacy with baricitinib, a marketed JAK1/JAK2 inhibitor. CONCLUSION: Our findings suggest that the novel JAK3/JAK1/TBK1 inhibitor CS12192 is potentially to alleviate autoimmune dermatosis.


Subject(s)
Disease Models, Animal , Janus Kinase 1 , Janus Kinase 3 , Protein Serine-Threonine Kinases , Sulfonamides , Animals , Mice , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 3/antagonists & inhibitors , Sulfonamides/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Female , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Purines/pharmacology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/immunology , Azetidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Psoriasis/drug therapy , Psoriasis/immunology , Psoriasis/pathology , Psoriasis/chemically induced , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/pathology , Pyrazoles
5.
Gene ; 927: 148719, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38917875

ABSTRACT

Renal cell carcinoma (RCC) represents a significant portion of genitourinary cancers, marked by challenging prognosis and high metastasis rates. Immunotherapy has been applied in managing advanced renal cell carcinoma, but the therapeutic outcomes are unsatisfactory. In this study, we order to construct a Janus kinase/signal transduction and activator transcriptional (JAK/STAT)-related signature linked to kidney patient outcomes for better predicting the efficacy to immune checkpoint inhibitors (ICIs) and to provide guidance for effective combination therapy. We screened 25 differentially expressed genes (DEGs) that exhibited high expression in RCC samples and were enriched in the JAK-STAT signaling pathway. Among these genes, 11 key genes were identified and correlated with the expectation of Kidney Clear Cell Carcinoma (KIRC) patients and all these genes was significantly elevated in RCC tumor tissues and cancer cells compared to para-cancer tissues and normal renal cells. Utilizing these 11 genes, we divided RCC patients into high-risk and low-risk groups. We found a clear correlation between the clinicopathologic factors of KIRC patients and the JAK-STAT-related risk score. And the IHC results shown that the JAK3 and STAT4 expression of tumor was significantly higher than normal tissue in RCC patients, the level of JAK3 and STAT4 was positively related to the T stage of RCC patients. In addition, high-risk patients had a poorer prognosis and greater protumor immune cell infiltration, and benefitted less from immunotherapy than did low-risk patients. Furthermore, the JAK-STAT-related risk score can predict disease-free survival (DFS) in RCC patients according to the nomogram, which constructed in combination with other clinical features such as age, TNM-staging and stage. Our study demonstrated the JAK-STAT signaling pathway's important regulatory function in RCC tumor immunity. This insight not only enhances our ability to accurately predict the survival rate of RCC patients, but also underscores a potential therapeutic alternative for RCC, involving the combined targeting of the JAK-STAT pathway and immune checkpoints.


Subject(s)
Biomarkers, Tumor , Carcinoma, Renal Cell , Gene Expression Regulation, Neoplastic , Immunotherapy , Kidney Neoplasms , Signal Transduction , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prognosis , Immunotherapy/methods , Female , Male , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/metabolism , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Janus Kinases/metabolism , Janus Kinases/genetics , Middle Aged , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Transcriptome , Gene Expression Profiling
6.
Bioorg Chem ; 149: 107499, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38815476

ABSTRACT

Janus Kinase 3 (JAK3) is important for the signaling transduction of cytokines in immune cells and is identified as potential target for treatment of rheumatoid arthritis (RA). Recently, we designed and synthesized two JAK3 inhibitors J1b and J1f, which featured with high selectivity but mild bioactivity. Therefore, in present study the structure was optimized to increase the potency. As shown in the results, most of the compounds synthesized showed stronger inhibitory activities against JAK3 in contrast to the lead compounds, among which 9a was the most promising candidate because it had the most potent effect in ameliorating carrageenan-induced inflammation of mice and exhibited low acute in vivo toxicity (MTD > 2 g/kg). Further analysis revealed that 9a was highly selective to JAK3 (IC50 = 0.29 nM) with only minimal effect on other JAK members (>3300-fold) and those kinases bearing a thiol in a position analogous to that of Cys909 in JAK3 (>150-fold). Meanwhile, the selectivity of JAK3 was also confirmed by PBMC stimulation assay, in which 9a irreversibly bound to JAK3 and robustly inhibited the signaling transduction with mild suppression on other JAKs. Moreover, it was showed that 9a could remarkably inhibited the proliferation of lymphocytes in response to concanavalin A and significantly mitigate disease severity in collagen induced arthritis. Therefore, present data indicate that compound 9a is a selective JAK3 inhibitor and could be a promising candidate for clinical treatment of RA.


Subject(s)
Arthritis, Rheumatoid , Janus Kinase 3 , Protein Kinase Inhibitors , Pyrimidines , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Arthritis, Rheumatoid/drug therapy , Animals , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Humans , Structure-Activity Relationship , Mice , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Molecular Structure , Dose-Response Relationship, Drug , Pyrroles/chemistry , Pyrroles/pharmacology , Pyrroles/chemical synthesis , Carrageenan , Male , Arthritis, Experimental/drug therapy , Arthritis, Experimental/chemically induced , Antirheumatic Agents/pharmacology , Antirheumatic Agents/chemistry , Antirheumatic Agents/chemical synthesis , Molecular Docking Simulation
7.
J Transl Med ; 22(1): 370, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637842

ABSTRACT

JAK-STAT signalling pathway inhibitors have emerged as promising therapeutic agents for the treatment of hair loss. Among different JAK isoforms, JAK3 has become an ideal target for drug discovery because it only regulates a narrow spectrum of γc cytokines. Here, we report the discovery of MJ04, a novel and highly selective 3-pyrimidinylazaindole based JAK3 inhibitor, as a potential hair growth promoter with an IC50 of 2.03 nM. During in vivo efficacy assays, topical application of MJ04 on DHT-challenged AGA and athymic nude mice resulted in early onset of hair regrowth. Furthermore, MJ04 significantly promoted the growth of human hair follicles under ex-vivo conditions. MJ04 exhibited a reasonably good pharmacokinetic profile and demonstrated a favourable safety profile under in vivo and in vitro conditions. Taken together, we report MJ04 as a highly potent and selective JAK3 inhibitor that exhibits overall properties suitable for topical drug development and advancement to human clinical trials.


Subject(s)
Drug Development , Hair , Mice , Animals , Humans , Mice, Nude , Drug Discovery , Janus Kinase 3
8.
J Clin Immunol ; 44(4): 98, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38598033

ABSTRACT

Biallelic null or hypomorphic variants in JAK3 cause SCID and less frequently Omenn syndrome. We investigated homozygous hypomorphic JAK3 mutations in two patients, and expression and function of a novel JAK3R431P variant in Omenn syndrome. Immunophenotyping of PBMC from the patient with the novel JAK3R431P variant was undertaken, by flow cytometry and Phosflow after stimulation with IL-2, IL-7, and IL-15. JAK3 expression was investigated by Western blotting. We report two patients with homozygous hypomorphic JAK3 variants and clinical features of Omenn syndrome. One patient had a previously described JAK3R775H variant, and the second had a novel JAK3R431P variant. One patient with a novel JAK3R431P variant had normal expression of JAK3 in immortalised EBV-LCL cells but reduced phosphorylation of STAT5 after stimulation with IL-2, IL-7, and IL-15 consistent with impaired kinase activity. These results suggest the JAK3R431P variant to be hypomorphic. Both patients are alive and well after allogeneic haematopoietic stem cell transplantation. They have full donor chimerism, restitution of thymopoiesis and development of appropriate antibody responses following vaccination. We expand the phenotype of hypomorphic JAK3 deficiency and demonstrate the importance of functional testing of novel variants in disease-causing genes.


Subject(s)
Janus Kinase 3 , Severe Combined Immunodeficiency , Humans , Infant , Interleukin-15 , Interleukin-2 , Interleukin-7 , Janus Kinase 3/genetics , Leukocytes, Mononuclear , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy
9.
Int J Mol Sci ; 25(5)2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38474223

ABSTRACT

The Janus kinase (JAK) family is a small group of protein tyrosine kinases that represent a central component of intracellular signaling downstream from a myriad of cytokine receptors. The JAK3 family member performs a particularly important role in facilitating signal transduction for a key set of cytokine receptors that are essential for immune cell development and function. Mutations that impact JAK3 activity have been identified in a number of human diseases, including somatic gain-of-function (GOF) mutations associated with immune cell malignancies and germline loss-of-function (LOF) mutations associated with immunodeficiency. The structure, function and impacts of both GOF and LOF mutations of JAK3 are highly conserved, making animal models highly informative. This review details the biology of JAK3 and the impact of its perturbation in immune cell-related diseases, including relevant animal studies.


Subject(s)
Immunologic Deficiency Syndromes , Neoplasms , Animals , Humans , Janus Kinase 3/metabolism , Signal Transduction , Janus Kinases/metabolism , Receptors, Cytokine/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism
10.
Int Immunopharmacol ; 132: 111931, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38547769

ABSTRACT

Peficitinib is a selective Janus kinase (JAK3) inhibitor recently developed and approved for the treatment of rheumatoid arthritis in Japan. Glycolysis in macrophages could induce NOD-like receptor (NLR) family and pyrin domain-containing protein 3 (NLRP3) inflammasome activation, thus resulting in pyroptosis and acute lung injury (ALI). The aim of our study was to investigate whether Peficitinib could alleviate lipopolysaccharide (LPS)-induced ALI by inhibiting NLRP3 inflammasome activation. Wild type C57BL/6J mice were intraperitoneally injected with Peficitinib (5 or 10 mg·kg-1·day-1) for 7 consecutive days before LPS injection. The results showed that Peficitinib pretreatment significantly relieved LPS-induced pulmonary edema, inflammation, and apoptosis. NLRP3 inflammasome and glycolysis in murine lung tissues challenged with LPS were also blocked by Peficitinib. Furthermore, we found that the activation of JAK3/signal transducer and activator of transcription 3 (STAT3) was also suppressed by Peficitinib in mice with ALI. However, in Jak3 knockout mice, Peficitinib did not show obvious protective effects after LPS injection. In vitro experiments further showed that Jak3 overexpression completely abolished Peficitinib-elicited inhibitory effects on pyroptosis and glycolysis in LPS-induced RAW264.7 macrophages. Finally, we unveiled that LPS-induced activation of JAK3/STAT3 was mediated by toll-like receptor 4 (TLR4) in RAW264.7 macrophages. Collectively, our study proved that Peficitinib could protect against ALI by blocking JAK3-mediated glycolysis and pyroptosis in macrophages, which may serve as a promising candidate against ALI in the future.


Subject(s)
Acute Lung Injury , Adamantane/analogs & derivatives , Glycolysis , Janus Kinase 3 , Lipopolysaccharides , Mice, Inbred C57BL , Niacinamide , Niacinamide/analogs & derivatives , STAT3 Transcription Factor , Signal Transduction , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Janus Kinase 3/metabolism , Janus Kinase 3/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Glycolysis/drug effects , Mice , Signal Transduction/drug effects , Male , Niacinamide/pharmacology , Niacinamide/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mice, Knockout , Acrylamides/pharmacology , Acrylamides/therapeutic use , Inflammasomes/metabolism , Pyroptosis/drug effects , Lung/pathology , Lung/drug effects , Lung/metabolism , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology
11.
Arch Pharm (Weinheim) ; 357(6): e2300753, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38442328

ABSTRACT

Selective inhibition of Janus kinase 3 (JAK3) is a promising strategy for the treatment of autoimmune diseases. Based on the discovery of a hydrophobic pocket unutilized between the lead compound RB1 and the JAK3 protein, a series of covalent JAK3 inhibitors were prepared by introducing various aromatic fragments to RB1. Among them, J1b (JAK3 IC50 = 7.2 nM, other JAKs IC50 > 1000 nM) stood out because of its low toxicity (MTD > 2 g/kg) and superior anti-inflammatory activity in Institute of Cancer Research mice. Moreover, the acceptable bioavailability (F% = 31.69%) ensured that J1b displayed excellent immune regulation in collagen-induced arthritis mice, whose joints in the high-dose group were almost recovered to a normal state. Given its clear kinase selectivity (Bmx IC50 = 539.9 nM, other Cys909 kinases IC50 > 1000 nM), J1b was nominated as a highly selective JAK3 covalent inhibitor, which could be used to safely treat arthritis and other autoimmune diseases.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Drug Design , Janus Kinase 3 , Protein Kinase Inhibitors , Animals , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Mice , Arthritis, Experimental/drug therapy , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Rheumatoid/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Mice, Inbred DBA , Humans , Dose-Response Relationship, Drug , Molecular Structure , Male , Molecular Docking Simulation
12.
J Pharmacol Exp Ther ; 389(1): 40-50, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38336380

ABSTRACT

B-cell acute lymphoblastic leukemia (B-ALL) is the most prevalent type of cancer in young children and is associated with high levels of reactive oxygen species (ROS). The antioxidant N-acetylcysteine (NAC) was tested for its ability to alter disease progression in a mouse model of B-ALL. Mb1-CreΔPB mice have deletions in genes encoding PU.1 and Spi-B in B cells and develop B-ALL at 100% incidence. Treatment of Mb1-CreΔPB mice with NAC in drinking water significantly reduced the frequency of CD19+ pre-B-ALL cells infiltrating the thymus at 11 weeks of age. However, treatment with NAC did not reduce leukemia progression or increase survival by a median 16 weeks of age. NAC significantly altered gene expression in leukemias in treated mice. Mice treated with NAC had increased frequencies of activating mutations in genes encoding Janus kinases 1 and 3. In particular, frequencies of Jak3 R653H mutations were increased in mice treated with NAC compared with control drinking water. NAC opposed oxidization of PTEN protein ROS in cultured leukemia cells. These results show that NAC alters leukemia progression in this mouse model, ultimately selecting for leukemias with high Jak3 R653H mutation frequencies. SIGNIFICANCE STATEMENT: In a mouse model of precursor B-cell acute lymphoblastic leukemia associated with high levels of reactive oxygen species, treatment with N-acetylcysteine did not delay disease progression but instead selected for leukemic clones with activating R653H mutations in Janus kinase 3.


Subject(s)
Drinking Water , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Mice , Animals , Child, Preschool , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Janus Kinases , Mutation Rate , Reactive Oxygen Species/metabolism , Precursor Cells, B-Lymphoid/metabolism , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Mutation , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Disease Progression
13.
Am J Clin Dermatol ; 25(2): 299-314, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38263353

ABSTRACT

BACKGROUND: The ALLEGRO phase 2a and 2b/3 studies demonstrated that ritlecitinib, an oral JAK3/TEC family kinase inhibitor, is efficacious at doses of ≥ 30 mg in patients aged ≥ 12 years with alopecia areata (AA). OBJECTIVE: The objective of this study was to evaluate the safety of ritlecitinib in an integrated analysis of four studies in AA. METHODS: Two cohorts were analyzed: a placebo-controlled and an all-exposure cohort. Proportions and study size-adjusted incidence rates (IRs) of adverse events (AEs) of interest and laboratory abnormalities are reported. RESULTS: In the placebo-controlled cohort (n = 881; median exposure: 169 days), the proportion of ritlecitinib-treated patients with AEs was 70.2-75.4% across doses versus 69.5% in the placebo group; serious AEs occurred in 0-3.2% versus 1.9% for the placebo. A total of 19 patients permanently discontinued due to AEs (5 while receiving the placebo). In the all-exposure cohort (n = 1294), median ritlecitinib exposure was 624 days [2091.7 total patient-years (PY)]. AEs were reported in 1094 patients (84.5%) and serious AEs in 57 (4.4%); 78 (6.0%) permanently discontinued due to AEs. The most common AEs were headache (17.7%; 11.9/100 PY), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positive test (15.5%; 9.8/100 PY), and nasopharyngitis (12.4%; 8.2/100 PY). There were two deaths (breast cancer and acute respiratory failure/cardiorespiratory arrest). Proportions (IRs) were < 0.1% (0.05/100 PY) for opportunistic infections, 1.5% (0.9/100 PY) for herpes zoster, 0.5% (0.3/100 PY) for malignancies (excluding nonmelanoma skin cancer), and 0.2% (0.1/100 PY) for major adverse cardiovascular events. CONCLUSIONS: Ritlecitinib is well tolerated with an acceptable safety profile up to 24 months in patients aged ≥ 12 years with AA (video abstract and graphical plain language summary available). TRIAL REGISTRIES: ClinicalTrials.gov: NCT02974868 (date of registration: 11/29/2016), NCT04517864 (08/18/2020), NCT03732807 (11/07/2018), and NCT04006457 (07/05/2019).


Subject(s)
Alopecia Areata , Antineoplastic Agents , Tryptamines , Humans , Alopecia Areata/drug therapy , Alopecia Areata/epidemiology , Carbazoles , Janus Kinase 3 , Protein Kinase Inhibitors/adverse effects , SARS-CoV-2 , Treatment Outcome
14.
Mol Biomed ; 5(1): 3, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38172378

ABSTRACT

The disruptor of telomeric silencing 1-like (DOT1L), a specific histone methyltransferase that catalyzed methylation of histone H3 on lysine 79, was associated with the pathogenesis of many diseases, but its role in peritoneal fibrosis remained unexplored. Here, we examined the role of DOT1L in the expression and activation of protein tyrosine kinases and development of peritoneal fibrosis. We found that a significant rise of DOT1L expression in the fibrotic peritoneum tissues from long-term PD patients and mice. Inhibition of DOT1L significantly attenuated the profibrotic phenotypic differentiation of mesothelial cells and macrophages, and alleviated peritoneal fibrosis. Mechanistically, RNA sequencing and proteomic analysis indicated that DOT1L was mainly involved in the processes of protein tyrosine kinase binding and extracellular matrix structural constituent in the peritoneum. Chromatin immunoprecipitation (ChIP) showed that intranuclear DOT1L guided H3K79me2 to upregulate EGFR in mesothelial cells and JAK3 in macrophages. Immunoprecipitation and immunofluorescence showed that extranuclear DOT1L could interact with EGFR and JAK3, and maintain the activated signaling pathways. In summary, DOT1L promoted the expression and activation of tyrosine kinases (EGFR in mesothelial cells and JAK3 in macrophages), promoting cells differentiate into profibrotic phenotype and thus peritoneal fibrosis. We provide the novel mechanism of dialysis-related peritoneal fibrosis (PF) and the new targets for clinical drug development. DOT1L inhibitor had the PF therapeutic potential.


Subject(s)
Histone-Lysine N-Methyltransferase , Peritoneal Fibrosis , Protein-Tyrosine Kinases , Animals , Female , Humans , Male , Mice , ErbB Receptors/metabolism , ErbB Receptors/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Janus Kinase 3/metabolism , Janus Kinase 3/genetics , Macrophages/metabolism , Macrophages/drug effects , Mice, Inbred C57BL , Peritoneal Fibrosis/pathology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
15.
Sci Adv ; 9(51): eadi3770, 2023 Dec 22.
Article in English | MEDLINE | ID: mdl-38117887

ABSTRACT

Steroids are the standard treatment for allergic airway inflammation in asthma, but steroid-refractory asthma poses a challenge. Group 2 innate lymphoid cells (ILC2s), such as T helper 2 (TH2) cells, produce key asthma-related type 2 cytokines. Recent insights from mouse and human studies indicate a potential connection between ILC2s and steroid-resistant asthma. Here, we highlight that lung ILC2s, rather than TH2 cells, can develop steroid resistance, allowing them to persist and maintain their disease-driving activity even during steroid treatment. The emergence of multipotent IL-5+IL-13+IL-17A+ ILC2s is associated with steroid-resistant ILC2s. The Janus kinase 3 (JAK3)/signal transducer and activator of transcription (STAT) 3, 5, and 6 pathways contribute to the acquisition of steroid-resistant ILC2s. The JAK3 inhibitor reduces ILC2 survival, proliferation, and cytokine production in vitro and ameliorates ILC2-driven Alternaria-induced asthma. Furthermore, combining a JAK3 inhibitor with steroids results in the inhibition of steroid-resistant asthma. These findings suggest a potential therapeutic approach for addressing this challenging condition in chronic asthma.


Subject(s)
Asthma , Janus Kinase Inhibitors , Humans , Animals , Mice , Immunity, Innate , Lymphocytes/metabolism , Asthma/drug therapy , Asthma/metabolism , Cytokines/metabolism , Inflammation , Steroids , Janus Kinase 3
16.
J Immunol Res ; 2023: 8924603, 2023.
Article in English | MEDLINE | ID: mdl-38106519

ABSTRACT

Natural killer (NK) cell is an essential cytotoxic lymphocyte in our innate immunity. Activation of NK cells is of paramount importance in defending against pathogens, suppressing autoantibody production and regulating other immune cells. Common gamma chain (γc) cytokines, including IL-2, IL-15, and IL-21, are defined as essential regulators for NK cell homeostasis and development. However, it is inconclusive whether γc cytokine-driven NK cell activation plays a protective or pathogenic role in the development of autoimmunity. In this study, we investigate and correlate the differential effects of γc cytokines in NK cell expansion and activation. IL-2 and IL-15 are mainly responsible for NK cell activation, while IL-21 preferentially stimulates NK cell proliferation. Blockade of Janus tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway by either JAK inhibitors or antibodies targeting γc receptor subunits reverses the γc cytokine-induced NK cell activation, leading to suppression of its autoimmunity-like phenotype in vitro. These results underline the mechanisms of how γc cytokines trigger autoimmune phenotype in NK cells as a potential target to autoimmune diseases.


Subject(s)
Autoimmune Diseases , Interleukin-2 , Humans , Interleukin-2/metabolism , Interleukin-15 , Cytokines/metabolism , Janus Kinases/metabolism , Killer Cells, Natural , Autoimmune Diseases/drug therapy , Janus Kinase 3
17.
Molecules ; 29(1)2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38202604

ABSTRACT

This work aimed to find new inhibitors of the CYP3A4 and JAK3 enzymes, which are significant players in autoimmune diseases such as rheumatoid arthritis. Advanced computer-aided drug design techniques, such as pharmacophore and 3D-QSAR modeling, were used. Two strong 3D-QSAR models were created, and their predictive power was validated by the strong correlation (R2 values > 80%) between the predicted and experimental activity. With an ROC value of 0.9, a pharmacophore model grounded in the DHRRR hypothesis likewise demonstrated strong predictive ability. Eight possible inhibitors were found, and six new inhibitors were designed in silico using these computational models. The pharmacokinetic and safety characteristics of these candidates were thoroughly assessed. The possible interactions between the inhibitors and the target enzymes were made clear via molecular docking. Furthermore, MM/GBSA computations and molecular dynamics simulations offered insightful information about the stability of the binding between inhibitors and CYP3A4 or JAK3. Through the integration of various computational approaches, this study successfully identified potential inhibitor candidates for additional investigation and efficiently screened compounds. The findings contribute to our knowledge of enzyme-inhibitor interactions and may help us create more effective treatments for autoimmune conditions like rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Humans , Cysteine , Cytochrome P-450 CYP3A , Molecular Docking Simulation , Arthritis, Rheumatoid/drug therapy , Molecular Dynamics Simulation , Janus Kinase 3
18.
Acta Physiologica Sinica ; (6): 874-882, 2019.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-781387

ABSTRACT

The present study was aimed to investigate the effect of Janus kinase 3 (JAK3) on the migration of breast cancer cells and the underlying mechanism. The expression of JAK3 in breast cancer MCF-7 cells was silenced by siRNA (siJAK3). The migration ability of MCF-7 cells was detected by scratch test. The activity of store-operated calcium channel (SOCC) was detected by fluorescence calcium imaging. The expression levels of Orai1 and STIM1, key molecules in the process of store-operated calcium entry (SOCE) were detected by Western blot and RT-PCR. The results showed that 2-APB, an inhibitor of SOCC, could inhibit the migration ability of MCF-7 cells. siJAK3 transfection significantly inhibited the migration ability of MCF-7 cells, decreased the activity of SOCC, and down-regulated mRNA and protein expression levels of Orai1 and Stim1. Over-expression of Orai1 or STIM1 in JAK3-silenced cells restored their migration ability. These results suggest that JAK3 facilitates the migration of breast cancer cells by SOCC.


Subject(s)
Humans , Breast Neoplasms , Calcium , Metabolism , Calcium Channels , Metabolism , Cell Movement , Physiology , Gene Expression Regulation, Neoplastic , Janus Kinase 3 , Genetics , Metabolism , MCF-7 Cells , ORAI1 Protein , Genetics
19.
Lima; IETSI; mar. 2018.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1362013

ABSTRACT

INTRODUCCIÓN: El presente dictamen expone la evaluación múltiple de abatacept, tofacitinib y adalimumab para el tratamiento de pacientes con artritis reumatoide (AR) activa moderada a severa con falla a anti-TNF y anti-CD20. El objetivo del tratamiento de la artritis reumatoide (AR) de inducir y mantener la remisión clínica para limitar la progresión del deterioro de las articulaciones, no es posible lograrlo con un solo tratamiento. El manejo clínico de esta enfermedad requiere disponer de varias opciones terapéuticas debido a su curso clínico crónico con actividad de enfermedad fluctuante y eventuales fallas a los tratamientos instaurados. En la institución se encuentran disponibles dos anti-TNF y rituximab para el manejo progresivo de los pacientes con AR activa. Sin embargo, algunos pacientes no responderán a estos tratamientos o eventualmente perderán la respuesta inicial, es por ello que se requiere evaluar otras opciones terapéuticas para estos pacientes. METODOLOGÍA: Se realizó una búsqueda de la literatura con respecto a la eficacia y seguridad comparativa entre abatacept, tofacitinib y adalimumab para el tratamiento de pacientes con artritis reumatoide (AR) moderada a severa con falla a un anti-TNF y un anti-CD20. Esta búsqueda se realizó utilizando los meta-buscadores: Translating Research into Practice (TRIPDATABASE), National Library of Medicine (Pubmed-Medline) y Health Systems Evidence. Adicionalmente, se amplió la búsqueda revisando la evidencia generada por grupos internacionales que realizan revisiones sistemáticas (RS), evaluación de tecnologías sanitarias (ETS) y guías de práctica clínica (GPC), tales como la Cochrane Group, The National Institute for Health and Care Excellence (NICE), the Agency for Health care Research and Quality (AHRQ), The Canadian Agency for Drugs and Technologies in Health (CADTH) y the Scottish Medicines Consortium (SMC). Esta búsqueda se completó ingresando a la página web www.clinicaltrials.gov, para así poder identificar ensayos clínicos en elaboración o que no hayan sido publicados aún, y asím disminuir el riesgo de sesgo de publicación. RESULTADOS: Se realizó la búsqueda bibliográfica y de evidencia científica respecto a la eficacia clínica relativa y perfil de toxicidad entre el uso de abatacept, tofacitinib y adalimumab en el tratamiento de pacientes con artritis reumatoide (AR) activa moderada a severa con falla a anti-TNF y anti-CD20. CONCLUSIONES: El objetivo del tratamiento de la artritis reumatoide es inducir y mantener la remisión clínica y limitar la progresión del deterioro de las articulaciones. El tratamiento debe ser lo más precoz posible después de realizado el diagnóstico a fin de obtener la remisión clínica. Las GPC consultadas no contienen recomendaciones específicas para los pacientes con AR que han fallado a un DMARD convencional, un anti-TNF y un anti-CD-20 (i.e. un convencional y dos biológicos). En general, las guías presentan recomendaciones de tratamiento homogéneas y claras hasta la falla de un DMARD convencional y un DMARD biológico, para quienes se recomienda usar un biológico del mismo o diferente mecanismo de acción. La ETS de NICE hace una recomendación condicionada a la aplicación del descuento acordado de manera confidencial, para el uso de tofacitinib en pacientes que han fallado al menos a un DMARD biológico y no puede recibir rituximab. La ETS de Francia concluyó que, aunque tofacitinib probablemente tenga un impacto, este sea pequeño debido a que existen otras opciones con las que no ha sido comparado y la preocupación por sus efectos tóxicos a largo plazo que incluyen riesgos de efectos adversos cardiovasculares, gastrointestinales y neoplasias malignas. En el caso de abatacept, NICE recomienda el uso en pacientes que han fallado al menos a un biológico anti-TNF, al igual que otros biológicos anti-TNF (adalimumab, infliximab y etanercept), y solamente si el paciente no podía recibir rituximab debido a un evento adverso u otra contraindicación. Tofacitinib, abatacept no han sido comparados directamente con adalimumab. Los ensayos identificados sobre abatacept y tofacitinib presentan problemas relacionados con las características de la población estudiada, los tiempos de evaluación y perfil de toxicidad a largo plazo. El estudio de abatacept incluyó a pacientes que fallaron a un solo anti-TNF resultando en una recomendación equivalente al grupo de anti-TNF. Tofacitinib mantuvo los grupos aleatorizados solo hasta el tercer mes, cuando la práctica habitual es evaluar la respuesta al sexto mes, teniendo que incurrir en métodos de imputación para calcular los efectos a mayor plazo, práctica que disminuye la confianza en los resultados. Además, existe preocupación por mayores riesgos cardiovasculares, gastrointestinales y cancerígenos. Adalimumab es un anti-TNF que tiene extensa experiencia de uso fuera y dentro de la institución. Se usa en el tratamiento de varias patologías incluyendo varias enfermedades reumatológicas, y puede ser una alternativa más en pacientes con AR activa que han fallado a varios biológicos, incluyendo anti-TNF y no anti-TNF. No hay información específica en la población de interés, pero estudios en población que fallaron a un metotrexate muestran que el tratamiento con adalimumab es similar a tofacitinib y abatacept. En la actualidad, no es posible establecer una jerarquía preferencial dentro de los medicamentos biológicos propuestos en vista de la ausencia de datos comparativos de eficacia y/o tolerancia. Sin embargo, existe evidencia que muestra que tofacitinib o abatacept son similares a adalimumab en pacientes con falla previa a metotrexate. Aunque se trata de evidencia indirecta, cabe señalar, que, de acuerdo con las recomendaciones, la preferencia es usar un anti-TNF como segunda y tercera línea (después de un DMARD convencional), debido a la experiencia clínica y el seguimiento de 15 años y la excelente eficacia estructural de esta clase terapéutica. Por lo expuesto, el Instituto de Evaluación de Tecnologías en Salud e Investigación IETSI aprueba el uso de adalimumab en combinación con metotrexate en pacientes con artritis reumatoides activa moderada a severa que han fallado a anti-TNF y un anti-CD20; según lo establecido en el Anexo N° 01. La vigencia del presente dictamen preliminar es de dos años, la continuación de dicha aprobación estará́ sujeta a los resultados obtenidos de los pacientes que reciban este tratamiento, a los reportes de seguridad que puedan surgir durante la farmacovigilancia activa, la nueva evidencia que pueda surgir en el tiempo y a un análisis farmacoeconómico.


Subject(s)
Humans , Arthritis, Rheumatoid/drug therapy , Antigens, CD20/adverse effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Adalimumab/therapeutic use , Abatacept/therapeutic use , Tumor Necrosis Factor Inhibitors/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL