Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 334
Filter
1.
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
2.
Molecules ; 28(15)2023 Aug 06.
Article in English | MEDLINE | ID: mdl-37570884

ABSTRACT

Rheumatoid arthritis (RA) remains one of the most prevalent autoimmune diseases worldwide. Janus kinase 3 (JAK3) is an essential enzyme for treating autoimmune diseases, including RA. Molecular modeling techniques play a crucial role in the search for new drugs by reducing time delays. In this study, the 3D-QSAR approach is employed to predict new JAK3 inhibitors. Two robust models, both field-based with R2 = 0.93, R = 0.96, and Q2 = 87, and atom-based with R2 = 0.94, R = 0.97, and Q2 = 86, yielded good results by identifying groups that may readily direct their interaction. A reliable pharmacophore model, DHRRR1, was provided in this work to enable the clear characterization of chemical features, leading to the design of 13 inhibitors with their pIC50 values. The DHRRR1 model yielded a validation result with a ROC value of 0.87. Five promising inhibitors were selected for further study based on an ADMET analysis of their pharmacokinetic properties and covalent docking (CovDock). Compared to the FDA-approved drug tofacitinib, the pharmaceutical features, binding affinity and stability of the inhibitors were analyzed through CovDock, 300 ns molecular dynamics simulations, free energy binding calculations and ADMET predictions. The results show that the inhibitors have strong binding affinity, stability and favorable pharmaceutical properties. The newly predicted molecules, as JAK3 inhibitors for the treatment of RA, are promising candidates for use as drugs.


Subject(s)
2-Aminopurine , Antirheumatic Agents , Computer-Aided Design , Drug Design , Janus Kinase 3 , Janus Kinase Inhibitors , 2-Aminopurine/analogs & derivatives , 2-Aminopurine/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/pharmacology , Janus Kinase 3/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Piperidines/chemistry , Piperidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Arthritis, Rheumatoid/drug therapy , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Pharmacophore
3.
Dermatology ; 239(5): 818-827, 2023.
Article in English | MEDLINE | ID: mdl-37643592

ABSTRACT

BACKGROUND: The keloid treatment is still a thorny and complicated clinical problem, especially in multiple keloids induced by wound, severe burn, ethnic background or cultural behaviors, or unexplained skin healing. Mainstream treatments have limited efficacy in treating multiple keloids. As no oral treatment with painlessness and convenience is available, oral treatment strategies should be formulated. OBJECTIVES: This study aimed to investigate the efficacy and therapeutic mechanism of oral tofacitinib in keloid patients. METHODS: We recruited the 7 patients with keloid scars and prescribed 5 mg of tofacitinib twice a day orally with a maximum follow-up of 12 weeks. The Patient and Observer Scar Assessment Scale (POSAS), the Vancouver scar scale (VSS), ANTERA 3D camera, and the DUB Skin Scanner 75 were used to assess the characteristics of the lesion. Immunohistochemistry was performed to evaluate collagen synthesis, proliferation, and relative molecular pathways. Moreover, the effects of tofacitinib were assessed on keloid fibroblast in vitro. RESULTS: After 12 weeks of oral tofacitinib, significant improvement in POSAS, VSS, and Dermatology Life Quality Index (DLQI) scores was observed (p < 0.05). The volume, lesion height, and dermis thickness of the keloid decreased (p < 0.05). Moreover, significant decreases in the expression of collagen I, Ki67, p-STAT 3, and p-SMAD2 were observed after 12 weeks of administration. In vitro experiments suggested that tofacitinib treatment inhibits fibroblast proliferation and collagen I synthesis via suppression of STAT3 and SMAD2 pathway. CONCLUSION: Tofacitinib, a new candidate oral drug for keloid, could reduce keloid lesion volume by inhibiting collagen synthesis and inhibiting fibroblast proliferation, and alleviate itch and pain to obtain a better life quality.


Subject(s)
Janus Kinase 3 , Keloid , Humans , Collagen , East Asian People , Janus Kinase 1 , Janus Kinase 3/antagonists & inhibitors , Keloid/pathology , Skin/pathology , Treatment Outcome
4.
Front Immunol ; 13: 955038, 2022.
Article in English | MEDLINE | ID: mdl-36203601

ABSTRACT

Alopecia areata (AA) is an autoimmune disease caused by T cell-mediated destruction of the hair follicle (HF). Therefore, approaches that effectively disrupt pathogenic T cell responses are predicted to have therapeutic benefit for AA treatment. T cells rely on the duality of T cell receptor (TCR) and gamma chain (γc) cytokine signaling for their development, activation, and peripheral homeostasis. Ifidancitinib is a potent and selective next-generation JAK1/3 inhibitor predicted to disrupt γc cytokine signaling. We found that Ifidancitinib robustly induced hair regrowth in AA-affected C3H/HeJ mice when fed with Ifidancitinib in chow diets. Skin taken from Ifidancitinib-treated mice showed significantly decreased AA-associated inflammation. CD44+CD62L- CD8+ T effector/memory cells, which are associated with the pathogenesis of AA, were significantly decreased in the peripheral lymphoid organs in Ifidancitinib-treated mice. We observed high expression of co-inhibitory receptors PD-1 on effector/memory CD8+ T cells, together with decreased IFN-γ production in Ifidancitinib-treated mice. Furthermore, we found that γc cytokines regulated T cell exhaustion. Taken together, our data indicate that selective induction of T cell exhaustion using a JAK inhibitor may offer a mechanistic explanation for the success of this treatment strategy in the reversal of autoimmune diseases such as AA.


Subject(s)
Alopecia Areata , Autoimmune Diseases , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Janus Kinase Inhibitors , Alopecia Areata/drug therapy , Animals , CD8-Positive T-Lymphocytes , Cytokines/therapeutic use , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Mice , Mice, Inbred C3H , Programmed Cell Death 1 Receptor
5.
Mol Cancer Ther ; 21(9): 1393-1405, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35732501

ABSTRACT

Terminal T-cell exhaustion poses a significant barrier to effective anticancer immunotherapy efficacy, with current drugs aimed at reversing exhaustion being limited. Recent investigations into the molecular drivers of T-cell exhaustion have led to the identification of chronic IL2 receptor (IL2R)-STAT5 pathway signaling in mediating T-cell exhaustion. We targeted the key downstream IL2R-intermediate JAK 3 using a clinically relevant highly specific JAK3-inhibitor (JAK3i; PF-06651600) that potently inhibited STAT5-phosphorylation in vitro. Whereas pulsed high-dose JAK3i administration inhibited antitumor T-cell effector function, low-dose chronic JAK3i significantly improved T-cell responses and decreased tumor load in mouse models of solid cancer. Low-dose JAK3i combined with cellular and peptide vaccine strategies further decreased tumor load compared with both monotherapies alone. Collectively, these results identify JAK3 as a novel and promising target for combination immunotherapy.


Subject(s)
Immunotherapy , Janus Kinase 3 , Neoplasms , T-Lymphocytes , Animals , Janus Kinase 3/antagonists & inhibitors , Mice , Neoplasms/therapy , Phosphorylation , Receptors, Interleukin-2/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes/immunology
6.
Int J Mol Sci ; 23(10)2022 May 13.
Article in English | MEDLINE | ID: mdl-35628248

ABSTRACT

JAK3 differs from other JAK family members in terms of tissue distribution and functional properties, making it a promising target for autoimmune disease treatment. However, due to the high homology of these family members, targeting JAK3 selectively is difficult. As a result, exploiting small changes or selectively boosting affinity within the ATP binding region to produce new tailored inhibitors of JAK3 is extremely beneficial. PubChem CID 137321159 was used as the lead inhibitor in this study to preserve the characteristic structure and to collocate it with the redesigned new parent core structure, from which a series of 1,7-dihydro-dipyrrolo [2,3-b:3',2'-e] pyridine derivatives were obtained using the backbone growth method. From the proposed compounds, 14 inhibitors of JAK3 were found based on the docking scoring evaluation. The RMSD and MM/PBSA methods of molecular dynamics simulations were also used to confirm the stable nature of this series of complex systems, and the weak protein−ligand interactions during the dynamics were graphically evaluated and further investigated. The results demonstrated that the new parent core structure fully occupied the hydrophobic cavity, enhanced the interactions of residues LEU828, VAL836, LYS855, GLU903, LEU905 and LEU956, and maintained the structural stability. Apart from this, the results of the analysis show that the binding efficiency of the designed inhibitors of JAK3 is mainly achieved by electrostatic and VDW interactions and the order of the binding free energy with JAK3 is: 8 (−70.286 kJ/mol) > 11 (−64.523 kJ/mol) > 6 (−51.225 kJ/mol) > 17 (−42.822 kJ/mol) > 10 (−40.975 kJ/mol) > 19 (−39.754 kJ/mol). This study may provide a valuable reference for the discovery of novel JAK3 inhibitors for those patients with immune diseases.


Subject(s)
Autoimmune Diseases , Janus Kinase 3 , Protein Kinase Inhibitors , Pyridines , Drug Design , Humans , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/chemistry , Janus Kinase 3/metabolism , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology
7.
Org Biomol Chem ; 20(7): 1453-1461, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35088800

ABSTRACT

The directing group assisted decarboxylative ortho-benzoylation of N-aryl-7-azaindoles with α-keto acids has been achieved by synergistic visible light promoted photoredox and palladium catalysis. The approach tenders rapid entry to aryl ketone architectures from simple α-keto acid precursors via the in situ generation of a benzoyl radical intermediate. The transformation provides a range of ortho-benzoylated N-aryl-7-azaindoles, with excellent site-selectivity and good functional group compatibility under mild reaction conditions. Biological target predictions indicate that these molecules may serve as potential anti-cancer and anti-viral agents.


Subject(s)
Enzyme Inhibitors/chemistry , Indoles/chemistry , Palladium/chemistry , Catalysis , Decarboxylation , Enzyme Inhibitors/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , Humans , Indoles/pharmacology , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/metabolism , Keto Acids/chemistry , Light , Molecular Structure , Oxidation-Reduction , Photochemical Processes , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Tankyrases/antagonists & inhibitors , Tankyrases/metabolism
9.
SAR QSAR Environ Res ; 33(1): 23-34, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34915777

ABSTRACT

The idea of using ranked binding energies of residues and data fusion are presented here for the first time as a valuable tool to classify active and selective inhibitors. Selective inhibitors of JAK3 can inhibit inflammatory cytokine while preventing targeting other subtypes of JAK1 and JAK2. Herein, we report a novel way to identify both active JAK3 and selective JAK1/JAK3 and JAK2/JAK3 inhibitors using the effective activity and selectivity classifications. The most important residues (top 10) responsible for the inhibition mechanism are sorted from high to low energies, which are considered as variables in the classification process. In addition, the ranked energies of ligands' heteroatoms (top 5), ranked energies of hydrogen bonds (top 5) and important molecular descriptors (top 10) were used to construct different data fusion possibilities. It is shown that the proposed data fusion strategy can increase the accuracy of the activity classification to 100% and the selectivity classification to 96.4%. The proposed strategies represented in this paper can help medicinal or pharmaceutical chemist in evaluation of both active and selective inhibitors before synthesizing new pharmaceuticals.


Subject(s)
Janus Kinase 3 , Protein Kinase Inhibitors , Pyrimidines , Quantitative Structure-Activity Relationship , Hydrogen Bonding , Janus Kinase 1 , Janus Kinase 2 , Janus Kinase 3/antagonists & inhibitors , Ligands , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology
11.
Bioorg Med Chem ; 50: 116457, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34670167

ABSTRACT

An aromatic trifluoromethyl ketone moiety was characterized as a new warhead for covalently reversible kinase inhibitor design to target the non-catalytic cysteine residue. Potent and selective covalently reversible inhibitors of FGFR4 kinase were successfully designed and synthesized by utilizing this new warhead. The binding mode of a representative inhibitor was fully characterized by using multiple technologies including MALDI-TOF mass spectrometry, dialysis assay and X-ray crystallographic studies etc. This functional group was also successfully applied to discovery of a new JAK3 inhibitor, suggesting its potential application in designing other kinase inhibitors.


Subject(s)
Hydrocarbons, Fluorinated/pharmacology , Janus Kinase 3/antagonists & inhibitors , Ketones/pharmacology , Protein Kinase Inhibitors/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Hydrocarbons, Fluorinated/chemical synthesis , Hydrocarbons, Fluorinated/chemistry , Janus Kinase 3/metabolism , Ketones/chemical synthesis , Ketones/chemistry , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
12.
Bioorg Chem ; 116: 105330, 2021 11.
Article in English | MEDLINE | ID: mdl-34547646

ABSTRACT

A series of cyano-substituted 2,4-diarylaminopyrimidines was designed and synthesized as potent non-covalent JAK3 inhibitors. Among the derivatives synthesized, 9o (IC50 = 22.86 nM), 9 k (IC50 = 21.58 nM), and 9j (IC50 = 20.66 nM) demonstrated inhibitory potencies against JAK3 similar to the known JAK3 inhibitor tofacitinib (IC50 = 20.10 nM). Moreover, 9o displayed potent anti-proliferative activities against Raji and Ramos cells, with IC50 values of 0.9255 µM and 1.405 µM, respectively. In addition, 9o demonstrated low toxicity in normal HBE (human bronchial epithelial cells, IC50 > 10 µΜ) and L-02 (human liver cells, IC50 = 3.104 µΜ) cells. Analysis of the mode of action by flow cytometry indicated that 9o effectively arrested Raji cells at the G2/M phase. Taken together, these results suggested that 9o might be a promising candidate for development as a potential treatment for B-cell lymphoma.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Janus Kinase 3/antagonists & inhibitors , Lymphoma, B-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Janus Kinase 3/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
13.
Dermatol Online J ; 27(7)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34391330

ABSTRACT

Dermatitis herpetiformis (DH) is a rare autoimmune blistering disorder in which patients with celiac disease, a gluten-sensitive enteropathy, present with a severely pruritic papulovesicular eruption over extensor surfaces such as the knees, elbows, lower back, buttocks, and neck. Patients are instructed to adhere to a gluten-free diet for purposes of improving their skin disease and gluten-sensitive enteropathy; this is the only treatment that lowers risk of enteropathy-associated T cell lymphoma. Patients who adhere to a strict gluten-free diet often have remission of their skin disease over months to years. Dapsone is a rapid and extremely effective first-line treatment option and often used while transitioning to a gluten-free diet. Aside from gluten-free diet and dapsone, second-line treatment options include sulfapyridine, sulfasalazine, and colchicine. Some patients have difficulty adhering to a gluten-free diet or develop intolerable side effects to systemic therapies. Furthermore, there is limited data on the use of the second-line treatments. Recent studies have shed light on the role of JAK-STAT-dependent pathways in the pathogenesis of dermatitis herpetiformis. We present a patient treated with tofacitinib, 5mg twice daily, an oral JAK1/3 inhibitor, who demonstrated clinical improvement of DH and control of new lesion development.


Subject(s)
Celiac Disease/complications , Dermatitis Herpetiformis/drug therapy , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Aged , Celiac Disease/diet therapy , Dapsone/therapeutic use , Dermatitis Herpetiformis/diet therapy , Dermatitis Herpetiformis/etiology , Dermatitis Herpetiformis/pathology , Diet, Gluten-Free , Drug Administration Schedule , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Male , Patient Compliance , Remission Induction/methods , Treatment Outcome
14.
Molecules ; 26(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34361750

ABSTRACT

The purpose of this work is to investigate the protein kinase inhibitory activity of constituents from Acacia auriculiformis stem bark. Column chromatography and NMR spectroscopy were used to purify and characterize betulin from an ethyl acetate soluble fraction of acacia bark. Betulin, a known inducer of apoptosis, was screened against a panel of 16 disease-related protein kinases. Betulin was shown to inhibit Abelson murine leukemia viral oncogene homolog 1 (ABL1) kinase, casein kinase 1ε (CK1ε), glycogen synthase kinase 3α/ß (GSK-3 α/ß), Janus kinase 3 (JAK3), NIMA Related Kinase 6 (NEK6), and vascular endothelial growth factor receptor 2 kinase (VEGFR2) with activities in the micromolar range for each. The effect of betulin on the cell viability of doxorubicin-resistant K562R chronic myelogenous leukemia cells was then verified to investigate its putative use as an anti-cancer compound. Betulin was shown to modulate the mitogen-activated protein (MAP) kinase pathway, with activity similar to that of imatinib mesylate, a known ABL1 kinase inhibitor. The interaction of betulin and ABL1 was studied by molecular docking, revealing an interaction of the inhibitor with the ABL1 ATP binding pocket. Together, these data demonstrate that betulin is a multi-target inhibitor of protein kinases, an activity that can contribute to the anticancer properties of the natural compound and to potential treatments for leukemia.


Subject(s)
Acacia/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Triterpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Apoptosis/genetics , Binding Sites , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase 1 epsilon/genetics , Casein Kinase 1 epsilon/metabolism , Cell Proliferation/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , K562 Cells , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , NIMA-Related Kinases/antagonists & inhibitors , NIMA-Related Kinases/genetics , NIMA-Related Kinases/metabolism , Plant Bark/chemistry , Plant Extracts/chemistry , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Triterpenes/chemistry , Triterpenes/isolation & purification , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
N Engl J Med ; 385(5): 406-415, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34133856

ABSTRACT

BACKGROUND: The efficacy and safety of tofacitinib, a Janus kinase inhibitor, in patients who are hospitalized with coronavirus disease 2019 (Covid-19) pneumonia are unclear. METHODS: We randomly assigned, in a 1:1 ratio, hospitalized adults with Covid-19 pneumonia to receive either tofacitinib at a dose of 10 mg or placebo twice daily for up to 14 days or until hospital discharge. The primary outcome was the occurrence of death or respiratory failure through day 28 as assessed with the use of an eight-level ordinal scale (with scores ranging from 1 to 8 and higher scores indicating a worse condition). All-cause mortality and safety were also assessed. RESULTS: A total of 289 patients underwent randomization at 15 sites in Brazil. Overall, 89.3% of the patients received glucocorticoids during hospitalization. The cumulative incidence of death or respiratory failure through day 28 was 18.1% in the tofacitinib group and 29.0% in the placebo group (risk ratio, 0.63; 95% confidence interval [CI], 0.41 to 0.97; P = 0.04). Death from any cause through day 28 occurred in 2.8% of the patients in the tofacitinib group and in 5.5% of those in the placebo group (hazard ratio, 0.49; 95% CI, 0.15 to 1.63). The proportional odds of having a worse score on the eight-level ordinal scale with tofacitinib, as compared with placebo, was 0.60 (95% CI, 0.36 to 1.00) at day 14 and 0.54 (95% CI, 0.27 to 1.06) at day 28. Serious adverse events occurred in 20 patients (14.1%) in the tofacitinib group and in 17 (12.0%) in the placebo group. CONCLUSIONS: Among patients hospitalized with Covid-19 pneumonia, tofacitinib led to a lower risk of death or respiratory failure through day 28 than placebo. (Funded by Pfizer; STOP-COVID ClinicalTrials.gov number, NCT04469114.).


Subject(s)
COVID-19 Drug Treatment , Glucocorticoids/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Antiviral Agents/therapeutic use , Brazil , COVID-19/complications , COVID-19/mortality , COVID-19/therapy , Double-Blind Method , Drug Therapy, Combination , Female , Hospitalization , Humans , Incidence , Janus Kinase 3/antagonists & inhibitors , Janus Kinase Inhibitors/adverse effects , Male , Middle Aged , Oxygen Inhalation Therapy , Piperidines/adverse effects , Pyrimidines/adverse effects , Respiratory Insufficiency/epidemiology , Respiratory Insufficiency/etiology
16.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33830087

ABSTRACT

The Janus kinase/signal transducers and activators of transcription (JAK/STAT) are key intracellular mediators in the signal transduction of many cytokines and growth factors. Common γ chain cytokines and interferon-γ that use the JAK/STAT pathway to induce biological responses have been implicated in the pathogenesis of alopecia areata (AA), a T cell-mediated autoimmune disease of the hair follicle. We previously showed that therapeutic targeting of JAK/STAT pathways using the first-generation JAK1/2 inhibitor, ruxolitinib, and the pan-JAK inhibitor, tofacitinib, was highly effective in the treatment of human AA, as well as prevention and reversal of AA in the C3H/HeJ mouse model. To better define the role of individual JAKs in the pathogenesis of AA, in this study, we tested and compared the efficacy of several next-generation JAK-selective inhibitors in the C3H/HeJ mouse model of AA, using both systemic and topical delivery. We found that JAK1-selective inhibitors as well as JAK3-selective inhibitors robustly induced hair regrowth and decreased AA-associated inflammation, whereas several JAK2-selective inhibitors failed to restore hair growth in treated C3H/HeJ mice with AA. Unlike JAK1, which is broadly expressed in many tissues, JAK3 expression is largely restricted to hematopoietic cells. Our study demonstrates inhibiting JAK3 signaling is sufficient to prevent and reverse disease in the preclinical model of AA.


Subject(s)
Alopecia Areata/drug therapy , Janus Kinase 3/metabolism , Protein Kinase Inhibitors/pharmacology , Administration, Topical , Alopecia Areata/metabolism , Alopecia Areata/prevention & control , Animals , Azetidines/administration & dosage , Azetidines/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Isonicotinic Acids/administration & dosage , Isonicotinic Acids/pharmacology , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Janus Kinase 3/antagonists & inhibitors , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C3H , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nitriles/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Triazoles/pharmacology
17.
Clin Exp Dermatol ; 46(5): 820-824, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33484582

ABSTRACT

Atopic dermatitis (AD) is chronic, pruritic, inflammatory skin disease that affects a significant portion of the population in industrialized nations. For nonresponders to conventional therapies, AD can significantly reduce sleep quality and quality of life. AD pathogenesis is multifactorial and involves multiple immune pathways, with recent evidence of T helper (Th)2, Th17 and Th22 axis attenuation in various AD endotypes and racial subtypes. Inhibition of the conserved Janus kinase (JAK) signalling pathway represents a promising therapeutic avenue to reduce the activation of multiple proinflammatory mediators involved in AD pathogenesis. JAK inhibitors exist in both oral and topical forms with variable specificity for the receptor tyrosine kinases JAK1, JAK2, JAK3 and tyrosine kinase 2. Oral formulations include abrocitinib, upadacitinib, baricitinib and gusacitinib, and are most appropriate for patients with moderate to severe AD. Emerging topical formulation in development include ruxolitinib and deglocitinib, which may be used in patients with localized AD and also adjunctively with systemic therapy in patients with more severe disease. With observed rapidity in itch relief and accompanying dramatic reduction in inflammatory lesion count, JAK inhibitors represent a promising new treatment to revolutionize the management of AD.


Subject(s)
Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Acetonitriles/administration & dosage , Acetonitriles/pharmacology , Acetonitriles/therapeutic use , Administration, Oral , Administration, Topical , Adult , Azetidines/administration & dosage , Azetidines/pharmacology , Azetidines/therapeutic use , Child , Dermatitis, Atopic/physiopathology , Dermatitis, Atopic/psychology , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Janus Kinase Inhibitors/administration & dosage , Nitriles/administration & dosage , Nitriles/pharmacology , Nitriles/therapeutic use , Piperidines/administration & dosage , Piperidines/pharmacology , Piperidines/therapeutic use , Purines/administration & dosage , Purines/pharmacology , Purines/therapeutic use , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridazines/administration & dosage , Pyridazines/pharmacology , Pyridazines/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quality of Life , STAT1 Transcription Factor/pharmacology , Safety , Severity of Illness Index , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , TYK2 Kinase/antagonists & inhibitors , Treatment Outcome
18.
Biochem Biophys Res Commun ; 558: 231-238, 2021 06 18.
Article in English | MEDLINE | ID: mdl-32113685

ABSTRACT

Several groups have developed in vitro expansion cultures for mouse metanephric nephron progenitor cells (NPCs) using cocktails of small molecules and growth factors including BMP7. However, the detailed mechanisms by which BMP7 acts in the NPC expansion remain to be elucidated. Here, by performing chemical screening for BMP substitutes, we identified a small molecule, TCS21311, that can replace BMP7 and revealed a novel inhibitory role of BMP7 in JAK3-STAT3 signaling in NPC expansion culture. Further, we found that TCS21311 facilitates the proliferation of mouse embryonic NPCs and human induced pluripotent stem cell-derived NPCs when added to the expansion culture. These results will contribute to understanding the mechanisms of action of BMP7 in NPC proliferation in vitro and in vivo and to the stable supply of NPCs for regenerative therapy, disease modeling and drug discovery for kidney diseases.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Janus Kinase Inhibitors/pharmacology , Nephrons/cytology , Nephrons/drug effects , Animals , Bone Morphogenetic Protein 7/administration & dosage , Cell Proliferation/drug effects , Cell Proliferation/physiology , Culture Media , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Janus Kinase 3/antagonists & inhibitors , Mice , Mice, 129 Strain , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Nephrons/metabolism , Signal Transduction/drug effects , Small Molecule Libraries
19.
Gastroenterol Hepatol ; 44(1): 39-48, 2021 Jan.
Article in English, Spanish | MEDLINE | ID: mdl-32829958

ABSTRACT

The use of Janus kinase (JAK) inhibitors is a new approach in the therapy of inflammatory diseases with immune base. Tofacitinib is one of these inhibitors targeting JAK1 and JAK3, and its efficacy has been demonstrated in the treatment of moderate to severe ulcerative colitis (UC). It is a small synthetic molecule administered orally, with a fast bioavailability and elimination rate, predictable pharmacokinetics and lack of immunogenicity, which are convenient characteristics for both efficacy and safety. This article reviews the pharmacological characteristics of tofacitinib and its safety profile.


Subject(s)
Colitis, Ulcerative/drug therapy , Janus Kinase Inhibitors/pharmacology , Piperidines/pharmacology , Pyrimidines/pharmacology , Arthritis, Rheumatoid , Drug Interactions , Herpes Zoster/chemically induced , Herpes Zoster Vaccine/administration & dosage , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 3/antagonists & inhibitors , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/pharmacokinetics , Neoplasms/immunology , Piperidines/adverse effects , Piperidines/pharmacokinetics , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Venous Thromboembolism/chemically induced
20.
Chem Biol Interact ; 333: 109316, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33285127

ABSTRACT

Streptomyces hygroscopicus UFPEDA 3370 was fermented in submerged cultivation and the biomass extract was partitioned, obtaining a fraction purified named EB1. After purification of EB1 fraction, nigericin free acid was obtained and identified. Nigericin presented cytotoxic activity against several cancer cell lines, being most active against HL-60 (human leukemia) and HCT-116 (human colon carcinoma) cell lines, presenting IC50 and (IS) values: 0.0014 µM, (30.0) and 0.0138 µM (3.0), respectively. On HCT-116, nigericin caused apoptosis and autophagy. In this study, nigericin was also screened both in vitro and in silico against a panel of cancer-related kinases. Nigericin was able to inhibit both JAK3 and GSK-3ß kinases in vitro and its binding affinities were mapped through the intermolecular interactions with each target in silico.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/pathology , Nigericin/pharmacology , Protein Kinase Inhibitors/pharmacology , Streptomyces/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Catalytic Domain , Cell Line, Tumor , Humans , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/chemistry , Janus Kinase 3/metabolism , Molecular Docking Simulation , Nigericin/chemistry , Nigericin/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...