1
|
Omar M, Omar M, Patt YS, Ukashi O, Sharif Y, Lahat A, Selinger CP, Sharif K. Genetic Risk of Ankylosing Spondylitis and Second-Line Therapy Need in Crohn's Disease: A Mendelian Randomization Study. J Clin Med 2024; 13:7496. [PMID: 39768419 PMCID: PMC11678710 DOI: 10.3390/jcm13247496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Crohn's disease (CD) and Ankylosing Spondylitis (AS) are chronic conditions with overlapping inflammatory pathways. This research investigates the genetic association between AS and the requirement for more aggressive therapeutic interventions in CD, suggesting a likelihood of increased severity in CD progression among individuals diagnosed with AS. Methods: This study utilized two-sample Mendelian randomization (TSMR) to analyze GWAS datasets for AS and CD requiring second-line treatment. Instrumental variables were selected based on single-nucleotide polymorphisms of genome-wide significance. Analytical methods included inverse-variance weighted (IVW), MR Egger, and other MR approaches, alongside sensitivity analysis, to validate the findings. Results: Our results indicated a significant association between AS genetic predisposition and the increased need for second-line treatments in CD. The IVW method showed an Odds Ratio (OR) of 2.16, and MR Egger provided an OR of 2.71, both were statistically significant. This association persisted even after the exclusion of influential outlier SNP rs2517655, confirming the robustness of our findings. Conclusions: This study suggests that genetic factors contributing to AS may influence the progression of CD, potentially necessitating more intensive treatment strategies. These findings underscore the importance of early screening in patients with co-existing AS and CD for tailoring treatment approaches, thus advancing personalized medicine in the management of these complex conditions.
Collapse
Affiliation(s)
- Mahmud Omar
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel;
| | - Mohammad Omar
- School of Medicine, V. N. Karazin Kharkiv National University, 61022 Kharkiv, Ukraine;
| | | | - Offir Ukashi
- Department of Gastroenterology, Sheba Medical Center, Tel-Hashomer 5262000, Israel; (O.U.); (A.L.)
| | - Yousra Sharif
- Department of Gastroenterology, Hadassah Medical Center, Jerusalem 91120, Israel;
| | - Adi Lahat
- Department of Gastroenterology, Sheba Medical Center, Tel-Hashomer 5262000, Israel; (O.U.); (A.L.)
| | | | - Kassem Sharif
- Internal Medicine B, Sheba Medical Centre, Ramat Gan 5262000, Israel;
- Department of Gastroenterology, Sheba Medical Center, Tel-Hashomer 5262000, Israel; (O.U.); (A.L.)
- Leeds Gastroenterology Institute, Leeds Teaching Hospitals, Leeds LS1 3EX, UK
| |
Collapse
|
2
|
Martins DM, Fernandes PO, Vieira LA, Maltarollo VG, Moraes AH. Structure-Guided Drug Design Targeting Abl Kinase: How Structure and Regulation Can Assist in Designing New Drugs. Chembiochem 2024; 25:e202400296. [PMID: 39008807 DOI: 10.1002/cbic.202400296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
The human protein Abelson kinase (Abl), a tyrosine kinase, plays a pivotal role in developing chronic myeloid leukemia (CML). Abl's involvement in various signaling pathways underscores its significance in regulating fundamental biological processes, including DNA damage responses, actin polymerization, and chromatin structural changes. The discovery of the Bcr-Abl oncoprotein, resulting from a chromosomal translocation in CML patients, revolutionized the understanding and treatment of the disease. The introduction of targeted therapies, starting with interferon-alpha and culminating in the development of tyrosine kinase inhibitors (TKIs) like imatinib, significantly improved patient outcomes. However, challenges such as drug resistance and side effects persist, indicating the necessity of research into novel therapeutic strategies. This review describes advancements in Abl kinase inhibitor development, emphasizing rational compound design from structural and regulatory information. Strategies, including bivalent inhibitors, PROTACs, and compounds targeting regulatory domains, promise to overcome resistance and minimize side effects. Additionally, leveraging the intricate structure and interactions of Bcr-Abl may provide insights into developing inhibitors for other kinases. Overall, this review highlights the importance of continued research into Abl kinase inhibition and its broader implications for therapeutic interventions targeting kinase-driven diseases. It provides valuable insights and strategies that may guide the development of next-generation therapies.
Collapse
MESH Headings
- Humans
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Drug Design
- Proto-Oncogene Proteins c-abl/metabolism
- Proto-Oncogene Proteins c-abl/antagonists & inhibitors
- Proto-Oncogene Proteins c-abl/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Molecular Structure
Collapse
Affiliation(s)
- Diego M Martins
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Philipe O Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Lucas A Vieira
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Vinícius G Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Adolfo H Moraes
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| |
Collapse
|
3
|
Golmohammadi M, Zamanian MY, Al‐Ani AM, Jabbar TL, Kareem AK, Aghaei ZH, Tahernia H, Hjazi A, Jissir SA, Hakimizadeh E. Targeting STAT3 signaling pathway by curcumin and its analogues for breast cancer: A narrative review. Animal Model Exp Med 2024; 7:853-867. [PMID: 39219410 PMCID: PMC11680487 DOI: 10.1002/ame2.12491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Breast cancer (BC) continues to be a significant global health issue, with a rising number of cases requiring ongoing research and innovation in treatment strategies. Curcumin (CUR), a natural compound derived from Curcuma longa, and similar compounds have shown potential in targeting the STAT3 signaling pathway, which plays a crucial role in BC progression. AIMS The aim of this study was to investigate the effects of curcumin and its analogues on BC based on cellular and molecular mechanisms. MATERIALS & METHODS The literature search conducted for this study involved utilizing the Scopus, ScienceDirect, PubMed, and Google Scholar databases in order to identify pertinent articles. RESULTS This narrative review explores the potential of CUR and similar compounds in inhibiting STAT3 activation, thereby suppressing the proliferation of cancer cells, inducing apoptosis, and inhibiting metastasis. The review demonstrates that CUR directly inhibits the phosphorylation of STAT3, preventing its movement into the nucleus and its ability to bind to DNA, thereby hindering the survival and proliferation of cancer cells. CUR also enhances the effectiveness of other therapeutic agents and modulates the tumor microenvironment by affecting tumor-associated macrophages (TAMs). CUR analogues, such as hydrazinocurcumin (HC), FLLL11, FLLL12, and GO-Y030, show improved bioavailability and potency in inhibiting STAT3, resulting in reduced cell proliferation and increased apoptosis. CONCLUSION CUR and its analogues hold promise as effective adjuvant treatments for BC by targeting the STAT3 signaling pathway. These compounds provide new insights into the mechanisms of action of CUR and its potential to enhance the effectiveness of BC therapies.
Collapse
Affiliation(s)
| | - Mohammad Yassin Zamanian
- Department of Physiology, School of MedicineHamadan University of Medical SciencesHamadanIran
- Department of Pharmacology and Toxicology, School of PharmacyHamadan University of Medical SciencesHamadanIran
| | - Ahmed Muzahem Al‐Ani
- Department of Medical Laboratories TechnologyAL‐Nisour University CollegeBaghdadIraq
| | | | - Ali Kamil Kareem
- Biomedical Engineering DepartmentAl‐Mustaqbal University CollegeHillahIraq
| | - Zeinab Hashem Aghaei
- Preventative Gynecology Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Hossein Tahernia
- Molecular Medicine Research Center, Research Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesPrince Sattam bin Abdulaziz UniversityAl‐KharjSaudi Arabia
| | | | - Elham Hakimizadeh
- Physiology‐Pharmacology Research Center, Research Institute of Basic Medical SciencesRafsanjan University of Medical SciencesRafsanjanIran
| |
Collapse
|
4
|
Mabry A, Kuzmicki CE, O'Brien A, Maggi LB, Weber JD. Elevated type I interferon signaling defines the proliferative advantage of ARF and p53 mutant tumor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623046. [PMID: 39605405 PMCID: PMC11601273 DOI: 10.1101/2024.11.11.623046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The tumor suppressors p53 and ARF collaborate to prevent unwarranted cell proliferation and as such are two of the most frequently mutated genes in human cancer. Concomitant loss of functional p53 and ARF leads to massive gains in cell proliferation and transformation and is often observed in some of the most aggressive human cancer subtypes. These phenotypic gains are preceded by increased type I interferon (IFN) signaling that involves canonical STAT1 activation and a subsequent IFN-stimulated gene (ISG) signature. Here, we show that cells lacking p53 and ARF require active JAK1 to phosphorylate STAT1 on Y701 to maintain their high rate of proliferation. In fact, the use of selective JAK1 inhibitors ruxolitinib or baricitinib inhibited the induction of ISG's and the proliferation of p53 and ARF deleted cells. We identify a group of solid human tumors that lack functional p53 and ARF, show an expression signature of the upregulated type I IFN response genes, and are sensitive to selective JAK1 inhibitors. These data suggest that the type I IFN response acts as a positive driver of proliferation in the absence of p53 and ARF and, as such, presents itself as a potential therapeutic target in aggressive solid tumors.
Collapse
|
5
|
Yuan J, Lu H, Zuo X, Yin L, Pu Y, Zhang J. Adverse Event Assessment of Upadacitinib: A Pharmacovigilance Study Based on the FAERS Database. Pharmacoepidemiol Drug Saf 2024; 33:e70030. [PMID: 39385705 DOI: 10.1002/pds.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE Upadacitinib, a Janus kinase (JAK) inhibitor, has been approved by the FDA to treat various autoimmune conditions. This study assessed its adverse events by analyzing reports from the FDA Adverse Event Reporting System (FAERS). METHODS FAERS data from Q3 2019 to Q4 2023 were extracted, and disproportionality analyses were conducted using four statistical measures, reporting odds ratio, proportionate reporting ratio, Bayesian confidence propagation neural network, and empirical Bayesian geometric mean. RESULTS A total of 6 879 398 adverse event reports were collected, with 37 700 reports identifying upadacitinib as the "primary suspected." These reports involved 24 system organ classes and 246 preferred terms that met the criteria across all four algorithms. The distribution of adverse events was assessed separately for female and male patients. Further analysis of the top 25 preferred terms revealed that, although the system organ classes were similar between sexes, the specific adverse events differed. The adverse events were analyzed by gender, showing musculoskeletal and skin disorders were prevalent and severe in male patients, while musculoskeletal issues, infections, and abnormal laboratory tests were common in female patients. Unexpected events like trigger finger, biliary sepsis, and serious events such as oral neoplasm were also identified. CONCLUSION This study provides real-world evidence for the safety evaluation of upadacitinib and underscores the need to monitor sex-specific adverse events. Future prospective studies are necessary to confirm these pharmacovigilance findings.
Collapse
Affiliation(s)
- Jiayu Yuan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - HongXia Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Xulei Zuo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
- Jiangsu Institute for Sport and Health (JISH), Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Gupta AO, Furcich JW, Nascene DR, Kemp S, King CJ, Nolan EE, Durose W, Miller BS, Orchard PJ, Lund TC. Targeting VEGF-mediated blood-brain barrier disruption in advanced cerebral leukodystrophy. J Neuroimmunol 2024; 393:578395. [PMID: 38897089 DOI: 10.1016/j.jneuroim.2024.578395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/13/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
The earliest clinical manifestation of cerebral adrenoleukodystrophy (CALD) is adrenal insufficiency (AI) characterized by elevations in ACTH and loss of cortisol. We showed high (though physiologically achievable) levels of ACTH increases endothelial permeability, increases anisotropy, and increases VEGF secretion. An ACBD1 knockout endothelial cell line had increased sensitivity to ACTH and VEGF. Inhibition of VEGF via application of anti-VEGF (bevacizumab) improved permeability. Six boys with advanced CALD were treated with bevacizumab combined with dexamethasone and ruxolitinib as immune suppressants. Most boys had decreases in gadolinium enhancement on MRI indicating improvement in endothelial function, though all boys continued to progress symptomatically.
Collapse
Affiliation(s)
- Ashish O Gupta
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Justin W Furcich
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - David R Nascene
- Department of Diagnostic Radiology, University of Minnesota Medical Center, Minneapolis, MN 55455, United States of America
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Carina J King
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Erin E Nolan
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Willa Durose
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Bradley S Miller
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Paul J Orchard
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America
| | - Troy C Lund
- Department of Pediatrics, Division of Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota Medical School, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
7
|
Wu H, Jiang F, Yuan W, Zhao Y, Liu N, Miao X. Rauwolfia polysaccharide can inhibit the progress of ulcerative colitis through NOS2-mediated JAK2/STAT3 pathway. PLoS One 2024; 19:e0301660. [PMID: 38626146 PMCID: PMC11020939 DOI: 10.1371/journal.pone.0301660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/20/2024] [Indexed: 04/18/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory disease of the digestive tract. Rauwolfia polysaccharide (Rau) has therapeutic effects on colitis in mice, but its mechanism of action needs to be further clarified. In the study, we explored the effect of Rau on the UC cell model induced by Lipopolysaccharide (LPS). METHODS We constructed a UC cell model by stimulating HT-29 cells with LPS. Dextran sodium sulfate (DSS) was used to induce mice to construct an animal model of UC. Subsequently, we performed Rau administration on the UC cell model. Then, the therapeutic effect of Rau on UC cell model and was validated through methods such as Cell Counting Kit-8 (CCK8), Muse, Quantitative real‑time polymerase chain reaction (RT-qPCR), Western blotting, and Enzyme-linked immunosorbent assay (ELISA). RESULTS The results showed that Rau can promote the proliferation and inhibit the apoptosis of the HT-29 cells-induced by LPS. Moreover, we observed that Rau can inhibit the expression of NOS2/JAK2/STAT3 in LPS-induced HT-29 cells. To further explore the role of NOS2 in UC progression, we used siRNA technology to knock down NOS2 and search for its mechanism in UC. The results illustrated that NOS2 knockdown can promote proliferation and inhibit the apoptosis of LPS-induced HT-29 cells by JAK2/STAT3 pathway. In addition, in vitro and in vivo experiments, we observed that the activation of the JAK2/STAT3 pathway can inhibit the effect of Rau on DSS-induced UC model. CONCLUSION In short, Rauwolfia polysaccharide can inhibit the progress of ulcerative colitis through NOS2-mediated JAK2/STAT3 pathway. This study provides a theoretical clue for the treatment of UC by Rau.
Collapse
Affiliation(s)
- Haidong Wu
- Department of Gastroenterology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Fan Jiang
- Medical Centre for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of the Center of Gerontology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Wei Yuan
- Department of Emergency Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Ye Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ning Liu
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xinpu Miao
- Department of Gastroenterology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
8
|
Zhang J, Wang D, Hu X. Estragole Ameliorates CFA Induced Rheumatoid Arthritis Symptoms in Wistar Rats by Inhibiting JAK-2/STAT-3 Pathway. Physiol Res 2024; 73:81-90. [PMID: 38466007 PMCID: PMC11019617 DOI: 10.33549/physiolres.935204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/17/2023] [Indexed: 04/26/2024] Open
Abstract
The present study was conducted to scrutinize the pharmacological effect of Estragole (ESG) against CFA-induced arthritis in rats. The rats underwent induction of arthritis using the administration of CFA and after that, the rats were randomly divided into five different groups, where three groups correspond to diverse dosages of ESG, and the other two were control and CFA-arthritic control. Results of the study suggested that ESG in a dose-dependent manner, improves body weight and arthritis score of rats as evidenced by reduction of hind-paw volume. ESG also improved the antioxidant status of rats by reducing MDA levels and enhancing the concentration of endogenous antioxidants SOD and GPx. The level of pro-inflammatory cytokines was also found to be reduced in the case of ESG treated group as compared to CFA-group. In a western blot analysis, ESH showed downregulation of p-JAK-2/STAT-3. The study provided concrete evidence for the protective effect of ESG against rheumatoid arthritis in rats.
Collapse
Affiliation(s)
- J Zhang
- Department of Rheumatology and Immunology, Zhangzhou Municipal Hospital Affiliated to Fujian Medical University, Zhangzhou, Fujian, China; Department of Orthopaedics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | | | | |
Collapse
|
9
|
Jiang H, Yang J, Li T, Wang X, Fan Z, Ye Q, Du Y. JAK/STAT3 signaling in cardiac fibrosis: a promising therapeutic target. Front Pharmacol 2024; 15:1336102. [PMID: 38495094 PMCID: PMC10940489 DOI: 10.3389/fphar.2024.1336102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
Cardiac fibrosis is a serious health problem because it is a common pathological change in almost all forms of cardiovascular diseases. Cardiac fibrosis is characterized by the transdifferentiation of cardiac fibroblasts (CFs) into cardiac myofibroblasts and the excessive deposition of extracellular matrix (ECM) components produced by activated myofibroblasts, which leads to fibrotic scar formation and subsequent cardiac dysfunction. However, there are currently few effective therapeutic strategies protecting against fibrogenesis. This lack is largely because the molecular mechanisms of cardiac fibrosis remain unclear despite extensive research. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling cascade is an extensively present intracellular signal transduction pathway and can regulate a wide range of biological processes, including cell proliferation, migration, differentiation, apoptosis, and immune response. Various upstream mediators such as cytokines, growth factors and hormones can initiate signal transmission via this pathway and play corresponding regulatory roles. STAT3 is a crucial player of the JAK/STAT pathway and its activation is related to inflammation, malignant tumors and autoimmune illnesses. Recently, the JAK/STAT3 signaling has been in the spotlight for its role in the occurrence and development of cardiac fibrosis and its activation can promote the proliferation and activation of CFs and the production of ECM proteins, thus leading to cardiac fibrosis. In this manuscript, we discuss the structure, transactivation and regulation of the JAK/STAT3 signaling pathway and review recent progress on the role of this pathway in cardiac fibrosis. Moreover, we summarize the current challenges and opportunities of targeting the JAK/STAT3 signaling for the treatment of fibrosis. In summary, the information presented in this article is critical for comprehending the role of the JAK/STAT3 pathway in cardiac fibrosis, and will also contribute to future research aimed at the development of effective anti-fibrotic therapeutic strategies targeting the JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Yu T, Yan J, Wang R, Zhang L, Hu X, Xu J, Li F, Sun Q. Integrative Multiomics Profiling Unveils the Protective Function of Ulinastatin against Dextran Sulfate Sodium-Induced Colitis. Antioxidants (Basel) 2024; 13:214. [PMID: 38397811 PMCID: PMC10886110 DOI: 10.3390/antiox13020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Ulcerative colitis is an inflammatory bowel disease with multiple pathogeneses. Here, we aimed to study the therapeutic role of ulinastatin (UTI), an anti-inflammatory bioagent, and its associated mechanisms in treating colitis. Dextran sulfate sodium was administrated to induce colitis in mice, and a subgroup of colitis mice was treated with UTI. The gut barrier defect and inflammatory manifestations of colitis were determined via histological and molecular experiments. In addition, transcriptomics, metagenomics, and metabolomics were employed to explore the possible mechanisms underlying the effects of UTI. We found that UTI significantly alleviated the inflammatory manifestations and intestinal barrier damage in the mice with colitis. Transcriptome sequencing revealed a correlation between the UTI treatment and JAK-STAT signaling pathway. UTI up-regulated the expression of SOCS1, which subsequently inhibited the phosphorylation of JAK2 and STAT3, thus limiting the action of inflammatory mediators. In addition, 16S rRNA sequencing illustrated that UTI maintained a more stable intestinal flora, protecting the gut from dysbiosis in colitis. Moreover, metabolomics analysis demonstrated that UTI indeed facilitated the production of some bile acids and short-chain fatty acids, which supported intestinal homeostasis. Our data provide evidence that UTI is effective in treating colitis and support the potential use of UTI treatment for patients with ulcerative colitis.
Collapse
Affiliation(s)
- Tianyu Yu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (T.Y.); (J.Y.); (L.Z.); (X.H.)
| | - Jun Yan
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (T.Y.); (J.Y.); (L.Z.); (X.H.)
| | - Ruochen Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China;
| | - Lei Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (T.Y.); (J.Y.); (L.Z.); (X.H.)
| | - Xiake Hu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (T.Y.); (J.Y.); (L.Z.); (X.H.)
| | - Jiaxi Xu
- Department of Physiology and Pathophysiology, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China;
| | - Fanni Li
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Qi Sun
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (T.Y.); (J.Y.); (L.Z.); (X.H.)
| |
Collapse
|
11
|
Sk MF, Samanta S, Poddar S, Kar P. Deciphering the molecular choreography of Janus kinase 2 inhibition via Gaussian accelerated molecular dynamics simulations: a dynamic odyssey. J Comput Aided Mol Des 2024; 38:8. [PMID: 38324213 DOI: 10.1007/s10822-023-00548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/24/2023] [Indexed: 02/08/2024]
Abstract
The Janus kinases (JAK) are crucial targets in drug development for several diseases. However, accounting for the impact of possible structural rearrangements on the binding of different kinase inhibitors is complicated by the extensive conformational variability of their catalytic kinase domain (KD). The dynamic KD contains mainly four prominent mobile structural motifs: the phosphate-binding loop (P-loop), the αC-helix within the N-lobe, the Asp-Phe-Gly (DFG) motif, and the activation loop (A-loop) within the C-lobe. These distinct structural orientations imply a complex signal transmission path for regulating the A-loop's flexibility and conformational preference for optimal JAK function. Nevertheless, the precise dynamical features of the JAK induced by different types of inhibitors still remain elusive. We performed comparative, microsecond-long, Gaussian accelerated molecular dynamics simulations in triplicate of three phosphorylated JAK2 systems: the KD alone, type-I ATP-competitive inhibitor (CI) bound KD in the catalytically active DFG-in conformation, and the type-II inhibitor (AI) bound KD in the catalytically inactive DFG-out conformation. Our results indicate significant conformational variations observed in the A-loop and αC helix motions upon inhibitor binding. Our studies also reveal that the DFG-out inactive conformation is characterized by the closed A-loop rearrangement, open catalytic cleft of N and C-lobe, the outward movement of the αC helix, and open P-loop states. Moreover, the outward positioning of the αC helix impacts the hallmark salt bridge formation between Lys882 and Glu898 in an inactive conformation. Finally, we compared their ligand binding poses and free energy by the MM/PBSA approach. The free energy calculations suggested that the AI's binding affinity is higher than CI against JAK2 due to an increased favorable contribution from the total non-polar interactions and the involvement of the αC helix. Overall, our study provides the structural and energetic insights crucial for developing more promising type I/II JAK2 inhibitors for treating JAK-related diseases.
Collapse
Affiliation(s)
- Md Fulbabu Sk
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India
- Theoretical and Computational Biophysics Group, Beckman Institute for Advanced Science and Technology, NIH Resource for Macromolecular Modeling and Visualization, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sunanda Samanta
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India
| | - Sayan Poddar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, MP, 453552, India.
| |
Collapse
|
12
|
Roskoski R. Properties of FDA-approved small molecule protein kinase inhibitors: A 2024 update. Pharmacol Res 2024; 200:107059. [PMID: 38216005 DOI: 10.1016/j.phrs.2024.107059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/14/2024]
Abstract
Owing to the dysregulation of protein kinase activity in many diseases including cancer, this enzyme family has become one of the most important drug targets in the 21st century. There are 80 FDA-approved therapeutic agents that target about two dozen different protein kinases and seven of these drugs were approved in 2023. Of the approved drugs, thirteen target protein-serine/threonine protein kinases, four are directed against dual specificity protein kinases (MEK1/2), twenty block nonreceptor protein-tyrosine kinases, and 43 inhibit receptor protein-tyrosine kinases. The data indicate that 69 of these drugs are prescribed for the treatment of neoplasms. Six drugs (abrocitinib, baricitinib, deucravacitinib, ritlecitinib, tofacitinib, upadacitinib) are used for the treatment of inflammatory diseases (atopic dermatitis, rheumatoid arthritis, psoriasis, alopecia areata, and ulcerative colitis). Of the 80 approved drugs, nearly two dozen are used in the treatment of multiple diseases. The following seven drugs received FDA approval in 2023: capivasertib (HER2-positive breast cancer), fruquintinib (metastatic colorectal cancer), momelotinib (myelofibrosis), pirtobrutinib (mantle cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma), quizartinib (Flt3-mutant acute myelogenous leukemia), repotrectinib (ROS1-positive lung cancer), and ritlecitinib (alopecia areata). All of the FDA-approved drugs are orally effective with the exception of netarsudil, temsirolimus, and trilaciclib. This review summarizes the physicochemical properties of all 80 FDA-approved small molecule protein kinase inhibitors including the molecular weight, number of hydrogen bond donors/acceptors, polar surface area, potency, solubility, lipophilic efficiency, and ligand efficiency.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
13
|
Malik MS, Alshareef HF, Alfaidi KA, Ather H, Abduljaleel Z, Hussein EM, Moussa Z, Ahmed SA. Exploring the untapped pharmacological potential of imidazopyridazines. RSC Adv 2024; 14:3972-3984. [PMID: 38288152 PMCID: PMC10823362 DOI: 10.1039/d3ra07280k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/10/2024] [Indexed: 01/31/2024] Open
Abstract
Imidazopyridazines are fused heterocycles, like purines, with a pyridazine ring replacing the pyrimidine ring in purines. Imidazopyridazines have been primarily studied for their kinase inhibition activity in the development of new anticancer and antimalarial agents. In addition to this, they have also been investigated for their anticonvulsant, antiallergic, antihistamine, antiviral, and antitubercular properties. Herein, we review the background and development of different imidazopyridazines as potential pharmacological agents. Moreover, the scope of this relatively less charted heterocyclic scaffold is also highlighted.
Collapse
Affiliation(s)
- M Shaheer Malik
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Hossa F Alshareef
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Khalid A Alfaidi
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Hissana Ather
- Science and Technology Unit, Umm Al-Qura University Makkah 21955 Saudi Arabia
| | - Zainularifeen Abduljaleel
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University (KKU) Abha 62529 Saudi Arabia
| | - Essam M Hussein
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah 21955 Saudi Arabia
- Department of Chemistry, Faculty of Science, Assiut University 71516 Assiut Egypt
| | - Ziad Moussa
- Department of Chemistry, College of Science, United Arab Emirates University P.O. Box 15551 Al Ain United Arab Emirates
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah 21955 Saudi Arabia
- Department of Chemistry, Faculty of Science, Assiut University 71516 Assiut Egypt
| |
Collapse
|
14
|
Roskoski R. Cost in the United States of FDA-approved small molecule protein kinase inhibitors used in the treatment of neoplastic and non-neoplastic diseases. Pharmacol Res 2024; 199:107036. [PMID: 38096958 DOI: 10.1016/j.phrs.2023.107036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
Because genetic alterations including mutations, overexpression, translocations, and dysregulation of protein kinases are involved in the pathogenesis of many illnesses, this enzyme family is the target of many drug discovery programs worldwide. The FDA has approved 80 small molecule protein kinase inhibitors with 77 drugs orally bioavailable. The data indicate that 69 of these medicinals are approved for the management of neoplasms including solid tumors such as breast and lung cancer as well as non-solid tumors such as leukemia. Moreover, the remaining 11 drugs target non-neoplastic diseases including psoriasis, rheumatoid arthritis, and ulcerative colitis. The cost of drugs was obtained from www.pharmacychecker.com using the FDA label to determine the dosage and number of tablets required per day. This methodology excludes any private or governmental insurance coverage, which would cover the entire cost or more likely a fraction of the stated price. The average monthly cost for the treatment of neoplastic diseases was $17,900 with a price of $44,000 for futibatinib (used to treat cholangiocarcinomas with FGFR2 fusions) and minimum of $5100 for binimetinib (melanoma). The average monthly cost for the treatment of non-neoplastic diseases was $6800 with a maximum of $17,000 for belumosudil (graft vs. host disease) and a minimum of $200 for netarsudil eye drops (glaucoma). There is a negative correlation of the cost of the drugs and the incidence of the targeted disease. Many of these agents are or were designated as orphan drugs meaning that there are fewer than 200,000 potential patients in the United States.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
15
|
Du SS, Fang YQ, Zhang W, Rao GW. Targeting TYK2 for Fighting Diseases: Recent Advance of TYK2 Inhibitors. Curr Med Chem 2024; 31:2900-2920. [PMID: 38904160 DOI: 10.2174/0929867330666230324163414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/03/2023] [Accepted: 02/03/2023] [Indexed: 06/22/2024]
Abstract
TYK2 (tyrosine-protein kinase 2) is a non-receptor protein kinase belonging to the JAK family and is closely associated with various diseases, such as psoriasis, inflammatory bowel disease, systemic lupus erythematosus. TYK2 activates the downstream proteins STAT1-5 by participating in the signal transduction of immune factors such as IL-12, IL-23, and IL-10, resulting in immune expression. The activity of the inhibitor TYK2 can effectively block the transduction of excessive immune signals and treat diseases. TYK2 inhibitors are divided into two types of inhibitors according to the different binding sites. One is a TYK2 inhibitor that binds to JH2 and inhibits its activity through an allosteric mechanism. The representative inhibitor is BMS-986165, developed by Bristol-Myers Squibb. The other class binds to the JH1 adenosine triphosphate (ATP) site and prevents the catalytic activity of the kinase by blocking ATP and downstream phosphorylation. This paper mainly introduces the protein structure, signaling pathway, synthesis, structure-activity relationship and clinical research of TYK2 inhibitors.
Collapse
Affiliation(s)
- Si-Shi Du
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Yu-Qing Fang
- College of Pharmaceutical Science, Zhejiang University of Technology, and Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Wen Zhang
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou, 310014, P. R. China
| |
Collapse
|
16
|
Zhao Y, Mu Y, Zou Y, He Z, Lu T, Wang X, Li W, Gao B. Conjoint research of WGCNA, single-cell transcriptome and structural biology reveals the potential targets of IDD development and treatment and JAK3 involvement. Aging (Albany NY) 2023; 15:14764-14790. [PMID: 38095643 PMCID: PMC10781489 DOI: 10.18632/aging.205289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/02/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVES This study conducted integrated analysis of bulk RNA sequencing, single-cell RNA sequencing and Weighted Gene Co-expression Network Analysis (WGCNA), to comprehensively decode the most essential genes of intervertebral disc degeneration (IDD); then mainly focused on the JAK3 macromolecule to identify natural compounds to provide more candidate drug options in alleviating IDD. METHODS In the first part, we performed single-cell transcriptome analysis and WGCNA workflow to delineate the most pivotal genes of IDD. Then series of structural biology approaches and high-throughput virtual screening techniques were performed to discover potential compounds targeting JAK-STAT signaling pathway, such as Libdock, ADMET, precise molecular docking algorithm and in-vivo drug stability assessment. RESULTS Totally 4 hub genes were determined in the development of IDD, namely VEGFA, MMP3, TNFSF11, and TIMP3, respectively. Then, 3 novel natural materials, ZINC000014952116, ZINC000003938642 and ZINC000072131515, were determined as potential compounds, with less toxicities and moderate ADME characteristics. In-vivo drug stability assessment suggested that these drugs could interact with JAK3, and their ligand-JAK3 complexes maintained the homeostasis in-vivo, which acted as regulatory role to JAK3 protein. Among them, ZINC000072131515, also known as Menaquinone, demonstrated significant protective roles to alleviate the progression of IDD in vitro, which proved the nutritional therapy in alleviating IDD. CONCLUSIONS This study reported the essential genes in the development of IDD, and also the roles of Menaquinone to ameliorate IDD through inhibiting JAK3 protein. This study also provided more options and resources on JAK3 targeted screening, which may further expand the drug resources in the pharmaceutical market.
Collapse
Affiliation(s)
- Yingjing Zhao
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yuxue Mu
- Aerospace Clinical Medical Center, School of Aerospace Medicine, Air Force Medical University, Xi’an, China
| | - Yujia Zou
- Department of Cardiology, Xinhua Hospital affiliated to School of Medicine, Shanghai Jiaotong University, China
| | - Zhijian He
- Department of Sports Teaching and Research, Lanzhou University, Lanzhou, China
| | - Tianxing Lu
- Zonglian College, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinhui Wang
- Department of Oncology, The Fifth Affiliated Hospital of Xinxiang Medical College, Xin Xiang 453100, China
| | - Weihang Li
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
17
|
Zhang JY, Sun JF, Nie P, Herdewijn P, Wang YT. Synthesis and clinical application of small-molecule inhibitors of Janus kinase. Eur J Med Chem 2023; 261:115848. [PMID: 37793326 DOI: 10.1016/j.ejmech.2023.115848] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/06/2023]
Abstract
Janus kinase (JAK) plays a crucial role in intracellular signaling pathways, particularly in cytokine-mediated signal transduction, making them attractive therapeutic targets for a wide range of diseases, including autoimmune disorders, myeloproliferative neoplasms, and inflammatory conditions. The review provides a comprehensive overview of the development and therapeutic potential of small-molecule inhibitors targeting JAK family of proteins in various clinical trials. It also discusses the mechanisms of action, specificity, and selectivity of these inhibitors, shedding light on the challenges associated with achieving target selectivity while minimizing off-target effects. Moreover, the review offers insights into the clinical applications of JAK inhibitors, summarizing the ongoing clinical trials and the Food and Drug Administration (FDA)-approved JAK inhibitors currently available for various diseases. Overall, this review provides a thorough examination of the synthesis and clinical use of typical small-molecule JAK inhibitors in different clinical stages and offers a bright future for the development of novel small-molecule JAK inhibitors.
Collapse
Affiliation(s)
- Jing-Yi Zhang
- College of Chemistry and Chemical Engineering, Zhengzhou Normal University, Zhengzhou, 450044, China
| | - Jin-Feng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin, 133002, China.
| | - Peng Nie
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Ya-Tao Wang
- First People's Hospital of Shangqiu, Henan Province, Shangqiu, 476100, China; Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| |
Collapse
|
18
|
Taha M, Elazab ST, Abdelbagi O, Saati AA, Babateen O, Baokbah TAS, Qusty NF, Mahmoud ME, Ibrahim MM, Badawy AM. Phytochemical analysis of Origanum majorana L. extract and investigation of its antioxidant, anti-inflammatory and immunomodulatory effects against experimentally induced colitis downregulating Th17 cells. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116826. [PMID: 37348796 DOI: 10.1016/j.jep.2023.116826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Origanum majorana L. is a member of the Lamiaceae family and is commonly used in Egyptian cuisine as a seasoning and flavor enhancer. It is also recognized as a well-known traditional medicine in Egypt and is widely used for treating abdominal colic due to its antispasmodic properties. However, the protective effects of Origanum majorana L. against ulcerative colitis and its underlying mechanisms remain unclear. AIM OF THE STUDY This study aimed to identify the biologically active components present in methanol extracts of Origanum majorana L. using gas chromatography/mass spectrometry (GC/MS). Additionally, it aimed to investigate the therapeutic effects of these extracts on acetic acid-induced ulcerative colitis and elucidate the potential mechanisms involved. MATERIALS AND METHODS We conducted a GC-MS analysis of the methanolic extract obtained from Origanum majorana L. Thirty-two male rats were included in the study and divided into four experimental groups, with eight rats in each group: sham, UC, UC + O. majorana, and UC sulfasalazine. After euthanizing the rats, colon tissue samples were collected for gross and microscopic examinations, assessment of oxidative stress, and molecular evaluation. GC-MS analysis identified 15 components in the extracts. Pretreatment with O. majorana L. extract and sulfasalazine significantly improved the disease activity index (DAI) and resulted in notable improvements in macroscopic and microscopic colon findings. Additionally, both treatments demonstrated preventive effects against colonic oxidative damage by reducing the levels of malondialdehyde (MDA) and increasing the levels of the antioxidant systems superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH), which operate through the Nrf2/HO-1 signaling pathway. Moreover, these treatments downregulated the colonic inflammatory cascade by inhibiting NFκB, TNFα, IL-1β, IL6, IL23, IL17, COX-2, and iNOS, subsequently leading to downregulation of the JAK2/STAT3 signaling pathway and a decrease in the Th17 cell response. Furthermore, a reduction in the number of apoptotic epithelial cells that expressed caspase-3 was observed. CONCLUSION pretreatment with O. majorana L. extract significantly ameliorated acetic acid-induced ulcerative colitis. This effect could be attributed to the protective, antioxidant, anti-inflammatory, and anti-apoptotic properties of the extract.
Collapse
Affiliation(s)
- Medhat Taha
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt; Department of Anatomy, Al- Qunfudah Medical College, Umm Al-Qura University, Al-Qunfudhah, 28814, Saudi Arabia.
| | - Sara T Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Omer Abdelbagi
- Department of Pathology, Qunfudah Faculty of Medicine, Umm-Al-Qura University, Kingdom of Saudi Arabia, Makka, 24382, Saudi Arabia
| | - Abdullah A Saati
- Department of Community Medicine and Pilgrims Healthcare, Faculty of Medicine, Umm Al-Qura University, Makkah, 24382, Saudi Arabia
| | - Omar Babateen
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, 24382, Saudi Arabia
| | - Tourki A S Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Al-Qunfudah, 28814, Saudi Arabia
| | - Naeem F Qusty
- Medical Laboratories Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, 24382, Saudi Arabia
| | - Mohamed Ezzat Mahmoud
- Histology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, 34711, Egypt
| | - Mohie Mahmoud Ibrahim
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Alaa M Badawy
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
19
|
Xi Y, Wen R, Zhang R, Dong Q, Hou S, Zhang S. Genetic evidence supporting a causal role of Janus kinase 2 in prostate cancer: a Mendelian randomization study. Aging Male 2023; 26:2257300. [PMID: 37706641 DOI: 10.1080/13685538.2023.2257300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Janus kinase-2 (JAK2) inhibitors are now being tried in basic research and clinical practice in prostate cancer (PCa). However, the causal relationship between JAK2 and PCa has not been uniformly described. Here, we examined the cause-effect relation between JAK2 and PCa. METHODS Two-sample Mendelian randomization (MR) analysis of genetic variation data of JAK2, PCa from IEU OpenGWAS Project was performed by inverse variance weighted, MR-Egger, and weighted median. Cochran's Q heterogeneity test and MR-Egger multiplicity analysis were performed to normalize the MR analysis results to reduce the effect of bias on the results. RESULTS Five instrumental variables were identified for further MR analysis. Specifically, combining the inverse variance-weighted (OR: 1.0009, 95% CI: 1.0001-1.0015, p = 0.02) and weighted median (OR: 1.0009, 95% CI: 1.0000-1.0017, p = 0.03). Sensitivity analysis showed that there was no heterogeneity (p = 0.448) and horizontal multiplicity (p = 0.770) among the instrumental variables. CONCLUSIONS We found JAK2 was associated with the development of PCa and was a risk factor for PCa, which might be instructive for the use of JAK2 inhibitors in PCa patients.
Collapse
Affiliation(s)
- Yujia Xi
- Department of Urology, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, PR China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, PR China
| | - Rui Wen
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, PR China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, PR China
| | - Ran Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, PR China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, PR China
| | - Qirui Dong
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, PR China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, PR China
| | - Sijia Hou
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, PR China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, PR China
- Department of Neurology, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Shengxiao Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, PR China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, PR China
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
20
|
Liu J, Han X, Zhang T, Tian K, Li Z, Luo F. Reactive oxygen species (ROS) scavenging biomaterials for anti-inflammatory diseases: from mechanism to therapy. J Hematol Oncol 2023; 16:116. [PMID: 38037103 PMCID: PMC10687997 DOI: 10.1186/s13045-023-01512-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Inflammation is a fundamental defensive response to harmful stimuli, but the overactivation of inflammatory responses is associated with most human diseases. Reactive oxygen species (ROS) are a class of chemicals that are generated after the incomplete reduction of molecular oxygen. At moderate levels, ROS function as critical signaling molecules in the modulation of various physiological functions, including inflammatory responses. However, at excessive levels, ROS exert toxic effects and directly oxidize biological macromolecules, such as proteins, nucleic acids and lipids, further exacerbating the development of inflammatory responses and causing various inflammatory diseases. Therefore, designing and manufacturing biomaterials that scavenge ROS has emerged an important approach for restoring ROS homeostasis, limiting inflammatory responses and protecting the host against damage. This review systematically outlines the dynamic balance of ROS production and clearance under physiological conditions. We focus on the mechanisms by which ROS regulate cell signaling proteins and how these cell signaling proteins further affect inflammation. Furthermore, we discuss the use of potential and currently available-biomaterials that scavenge ROS, including agents that were engineered to reduce ROS levels by blocking ROS generation, directly chemically reacting with ROS, or catalytically accelerating ROS clearance, in the treatment of inflammatory diseases. Finally, we evaluate the challenges and prospects for the controlled production and material design of ROS scavenging biomaterials.
Collapse
Affiliation(s)
- Jiatong Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoyue Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Tingyue Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Keyue Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Zhaoping Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, China.
- Department of Prosthodontics, West China School of Stomatology, Sichuan University, No. 14, Section 3, Renmin Nanlu, Chengdu, 610041, China.
| |
Collapse
|
21
|
Fang Z, Sun H, Wang Y, Sun Z, Yin M. Discovery of WD-890: A novel allosteric TYK2 inhibitor for the treatment of multiple autoimmune diseases. Biomed Pharmacother 2023; 167:115611. [PMID: 37778274 DOI: 10.1016/j.biopha.2023.115611] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023] Open
Abstract
Tyrosine kinase 2 (TYK2) as a member of Janus kinase (JAK) family, mainly mediates the signaling of type I interferons (IFN), interleukin-12 (IL-12) and interleukin-23 (IL-23), which has become an attractive target for treatment of immune and inflammatory diseases. However, the development of selective TYK2 inhibitors is challenging due to the high homology of the catalytic kinase domain among the JAK family members. Here, we report a novel and potent allosteric inhibitor, WD-890, which binds to the pseudokinase domain of TYK2 with high selectivity and inhibits its function. We accomplished a series of preclinical studies to demonstrate the therapeutic efficacy of WD-890 in four animal models: systemic lupus erythematosus (SLE), psoriasis, psoriatic arthritis (PsA), and inflammatory bowel disease (IBD). The pharmacokinetic and toxicology results further indicate that WD-890 has favorable absorption, distribution, metabolism, and excretion (ADME) properties and tolerable toxicity. In conclusion, our study shows that WD-890 could be a promising oral TYK2 inhibitor for future treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Zhiqin Fang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongyin Sun
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China; Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Yutong Wang
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhenliang Sun
- Southern Medical University Affiliated Fengxian Hospital, Shanghai, China.
| | - Mingzhu Yin
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China; Translational Medicine Research Center (TMRC), School of Medicine Chongqing University, Shapingba, Chongqing, China.
| |
Collapse
|
22
|
Zhao C, Zhang Y, Zhang J, Li S, Liu M, Geng Y, Liu F, Chai Q, Meng H, Li M, Li J, Zheng Y, Zhang Y. Discovery of Novel Fedratinib-Based HDAC/JAK/BRD4 Triple Inhibitors with Remarkable Antitumor Activity against Triple Negative Breast Cancer. J Med Chem 2023; 66:14150-14174. [PMID: 37796543 DOI: 10.1021/acs.jmedchem.3c01242] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Multitarget HDAC inhibitors capable of simultaneously blocking the BRD4-LIFR-JAK1-STAT3 signaling pathway hold great potential for the treatment of TNBC and other solid tumors. Herein, novel Fedratinib-based multitarget HDAC inhibitors were rationally designed, synthesized, and biologically evaluated, among which compound 25ap stood out as a potent HDAC/JAK/BRD4 triple inhibitor. Satisfyingly, compound 25ap led to concurrent inhibition of HDACs and the BRD4-LIFR-JAK1-STAT3 signaling pathway, which was validated by hyper-acetylation of histone and α-tubulin, hypo-phosphorylation of STAT3, downregulation of LIFR, MCL-1, and c-Myc in MDA-MB-231 cells. The multitarget effects of 25ap contributed to its robust antitumor response, including potent antiproliferative activity, remarkable apoptosis-inducing activity, and inhibition of colony formation. Notably, 25ap possessed an acceptable therapeutic window between normal and cancerous cells, desirable in vitro metabolic stability in mouse microsome, and sufficient in vivo exposure via intraperitoneal administration. Additionally, the in vivo antitumor potency of 25ap was demonstrated in an MDA-MB-231 xenograft model.
Collapse
Affiliation(s)
- Chunlong Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Yu Zhang
- Key Lab of Advanced Drug Preparation Technologies (Ministry of Education), State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jin'ge Zhang
- Key Lab of Advanced Drug Preparation Technologies (Ministry of Education), State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Shunda Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Mengyang Liu
- Key Lab of Advanced Drug Preparation Technologies (Ministry of Education), State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yinping Geng
- Key Lab of Advanced Drug Preparation Technologies (Ministry of Education), State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Fengling Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Qipeng Chai
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Hongwei Meng
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Mengzhe Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Jintao Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| | - Yichao Zheng
- Key Lab of Advanced Drug Preparation Technologies (Ministry of Education), State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Henan Province for Drug Quality and Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yingjie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, P.R. China
| |
Collapse
|
23
|
Mahjoor M, Mahmoudvand G, Farokhi S, Shadab A, Kashfi M, Afkhami H. Double-edged sword of JAK/STAT signaling pathway in viral infections: novel insights into virotherapy. Cell Commun Signal 2023; 21:272. [PMID: 37784164 PMCID: PMC10544547 DOI: 10.1186/s12964-023-01240-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/19/2023] [Indexed: 10/04/2023] Open
Abstract
The Janus kinase/signal transducer and activator of transcription (JAK/STAT) is an intricate signaling cascade composed of various cytokines, interferons (IFN, growth factors, and other molecules. This pathway provides a delicate mechanism through which extracellular factors adjust gene expression, thereby acting as a substantial basis for environmental signals to influence cell growth and differentiation. The interactions between the JAK/STAT cascade and antiviral IFNs are critical to the host's immune response against viral microorganisms. Recently, with the emergence of therapeutic classes that target JAKs, the significance of this cascade has been recognized in an unprecedented way. Despite the functions of the JAK/STAT pathway in adjusting immune responses against viral pathogens, a vast body of evidence proposes the role of this cascade in the replication and pathogenesis of viral pathogens. In this article, we review the structure of the JAK/STAT signaling cascade and its role in immuno-inflammatory responses. We also highlight the paradoxical effects of this pathway in the pathogenesis of viral infections. Video Abstract.
Collapse
Affiliation(s)
- Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alireza Shadab
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
| | - Mojtaba Kashfi
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
| |
Collapse
|
24
|
Fan AC, Nakauchi Y, Bai L, Azizi A, Nuno KA, Zhao F, Köhnke T, Karigane D, Cruz-Hernandez D, Reinisch A, Khatri P, Majeti R. RUNX1 loss renders hematopoietic and leukemic cells dependent on IL-3 and sensitive to JAK inhibition. J Clin Invest 2023; 133:e167053. [PMID: 37581927 PMCID: PMC10541186 DOI: 10.1172/jci167053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
Disease-initiating mutations in the transcription factor RUNX1 occur as germline and somatic events that cause leukemias with particularly poor prognosis. However, the role of RUNX1 in leukemogenesis is not fully understood, and effective therapies for RUNX1-mutant leukemias remain elusive. Here, we used primary patient samples and a RUNX1-KO model in primary human hematopoietic cells to investigate how RUNX1 loss contributes to leukemic progression and to identify targetable vulnerabilities. Surprisingly, we found that RUNX1 loss decreased proliferative capacity and stem cell function. However, RUNX1-deficient cells selectively upregulated the IL-3 receptor. Exposure to IL-3, but not other JAK/STAT cytokines, rescued RUNX1-KO proliferative and competitive defects. Further, we demonstrated that RUNX1 loss repressed JAK/STAT signaling and rendered RUNX1-deficient cells sensitive to JAK inhibitors. Our study identifies a dependency of RUNX1-mutant leukemias on IL-3/JAK/STAT signaling, which may enable targeting of these aggressive blood cancers with existing agents.
Collapse
Affiliation(s)
- Amy C. Fan
- Immunology Graduate Program
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - Yusuke Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | | | - Armon Azizi
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
- University of California Irvine School of Medicine, Irvine, California, USA
| | - Kevin A. Nuno
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
- Cancer Biology Graduate Program, Stanford University School of Medicine, Stanford, California, USA
| | - Feifei Zhao
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - Thomas Köhnke
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - Daiki Karigane
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - David Cruz-Hernandez
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
- Medical Research Council (MRC) Molecular Haematology Unit and Oxford Centre for Haematology, University of Oxford, Oxford, United Kingdom
| | - Andreas Reinisch
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
- Division of Hematology, Medical University of Graz, Graz, Austria
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, School of Medicine, and
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine
- Cancer Institute
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
25
|
Lee YH, Song GG. Relative effectiveness and safety of interleukin-6 and Janus kinase inhibitors versus adalimumab in patients with rheumatoid arthritis: a network meta-analysis. Z Rheumatol 2023; 82:696-705. [PMID: 36427070 DOI: 10.1007/s00393-022-01290-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The relative efficacy and tolerability of interleukin‑6 (IL-6) and Janus kinase (JAK) inhibitor therapies were compared with those of adalimumab in patients with rheumatoid arthritis (RA) and inadequate responses to methotrexate (MTX). METHODS We performed a Bayesian network meta-analysis to combine direct and indirect evidence from randomized controlled trials (RCTs) to examine the efficacy and safety of IL‑6 inhibitors, JAK inhibitors, and adalimumab in patients with RA and inadequate responses to MTX. RESULTS Seven RCTs, which included 4428 patients (1066 for IL‑6 inhibitors and 3362 for JAK inhibitors), met the inclusion criteria. IL‑6 inhibitors were placed at the top left of the league table diagonal (Odds ratio, OR 1.43; 95% CrI 1.12-1.82) as these were correlated with the most beneficial ACR20 response rate. Conversely, the placebo was placed at the bottom right of the league table diagonal as it was correlated with the least desirable effects. IL‑6 and JAK inhibitors produced a substantial ACR20 response relative to adalimumab. The surface under the cumulative ranking curve (SUCRA) revealed that treatment with IL‑6 inhibitors had the greatest ability to reach the ACR20 response rate (SUCRA = 0.826), followed by treatment with JAK inhibitors (SUCRA = 0.672) and adalimumab (SUCRA = 0.001). The ACR50 and ACR70 rates displayed patterns similar to the ACR20 response rate. With regard to serious adverse events (SAEs), the SUCRA rating likelihood showed that adalimumab was likely to be the best intervention, followed by JAK and IL‑6 inhibitors. CONCLUSION Both IL‑6 and JAK inhibitors are more effective than adalimumab and have similar effects in patients with RA and an inadequate response to MTX. Adalimumab is likely to be safer than JAK and IL‑6 inhibitors.
Collapse
Affiliation(s)
- Young Ho Lee
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea (Republic of).
- Division of Rheumatology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, 73, Goryeodae-ro, 02841, Seongbuk-gu, Seoul, Korea (Republic of).
| | - Gwan Gyu Song
- Department of Rheumatology, Korea University College of Medicine, Seoul, Korea (Republic of)
| |
Collapse
|
26
|
Luan Y, Luan Y, He H, Jue B, Yang Y, Qin B, Ren K. Glucose metabolism disorder: a potential accomplice of SARS-CoV-2. Int J Obes (Lond) 2023; 47:893-902. [PMID: 37542197 DOI: 10.1038/s41366-023-01352-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023]
Abstract
Globally, 265,713,467 confirmed cases of SARS-CoV-2 (CoV-2), including 5,260,888 deaths, have been reported by the WHO. It is important to study the mechanism of this infectious disease. A variety of evidences show the potential association between CoV-2 and glucose metabolism. Notably, people with type 2 diabetes mellitus (T2DM) and other metabolic complications were prone to have a higher risk of developing a more severe infection course than people who were metabolically normal. The correlations between glucose metabolism and CoV-2 progression have been widely revealed. This review will discuss the association between glucose metabolism disorders and CoV-2 progression, showing the promoting effect of diabetes and other diseases related to glucose metabolism disorders on the progression of CoV-2. We will further conclude the effects of key proteins and pathways in glucose metabolism regulation on CoV-2 progression and potential interventions by targeting glucose metabolism disorders for CoV-2 treatment. Therefore, this review will provide systematic insight into the treatment of CoV-2 from the perspective of glucose metabolism.
Collapse
Affiliation(s)
- Yi Luan
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ying Luan
- State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100000, China
| | - Hongbo He
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Bolin Jue
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453000, China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Bo Qin
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
27
|
Hua H, Zhao Q, Xia J, Dai QL, Bai SR, Wang XB, Zhou M. Peficitinib ameliorates doxorubicin-induced cardiotoxicity by suppressing cellular senescence and enhances its antitumor activity. Int Immunopharmacol 2023; 122:110630. [PMID: 37451017 DOI: 10.1016/j.intimp.2023.110630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/28/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023]
Abstract
Irreversible cardiotoxicity limits the clinical applications of doxorubicin (DOX). Cardiotoxicity can be detected early using clinical assessment; however, effective preventive measures are still lacking. Peficitinib (ASP015K), a JAK (Janus kinase) inhibitor, is a potent anti-inflammatory agent in autoimmune diseases. Nevertheless, little research has been conducted on anti-ageing and anti-tumour therapies. In this study, we investigated whether ASP015K could attenuate DOX-induced cardiotoxicity through its anti-ageing effects and whether it would affect the tumour treatment effect of DOX by establishing senescence, acute heart injury, and xenograft models. We observed that ASP015K could antagonise the senescence induced by various factors, including hydrogen peroxide and DOX. In addition, ASP015K treatment significantly alleviated cardiac function damage, histopathological deterioration, myocardial fibrosis, and oxidative damage in acute injury mouse models. ASP015K enhanced the sensitivity of tumour cells to DOX therapy and significantly slowed down the tumour growth rate and tumour volume in the xenograft mouse model. Therefore, ASP015K is expected to be developed as a potential cardioprotective agent to prevent or reduce the cardiotoxic side effects of anthracyclines in chemotherapy.
Collapse
Affiliation(s)
- Hui Hua
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China; Key Laboratory of University Cell Biology Yunnan Province, Dali, Yunnan, 671000, China
| | - Qi Zhao
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China; Key Laboratory of University Cell Biology Yunnan Province, Dali, Yunnan, 671000, China
| | - Jing Xia
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China; Key Laboratory of University Cell Biology Yunnan Province, Dali, Yunnan, 671000, China
| | - Qian-Long Dai
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China; Key Laboratory of University Cell Biology Yunnan Province, Dali, Yunnan, 671000, China
| | - Shi-Rui Bai
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China; Key Laboratory of University Cell Biology Yunnan Province, Dali, Yunnan, 671000, China
| | - Xiao-Bo Wang
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China; Key Laboratory of University Cell Biology Yunnan Province, Dali, Yunnan, 671000, China.
| | - Min Zhou
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China.
| |
Collapse
|
28
|
Roskoski R. Small molecule protein kinase inhibitors approved by regulatory agencies outside of the United States. Pharmacol Res 2023; 194:106847. [PMID: 37454916 DOI: 10.1016/j.phrs.2023.106847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Owing to genetic alterations and overexpression, the dysregulation of protein kinases plays a significant role in the pathogenesis of many autoimmune and neoplastic disorders and protein kinase antagonists have become an important drug target. Although the efficacy of imatinib in the treatment of chronic myelogenous leukemia in the United States in 2001 was the main driver of protein kinase inhibitor drug discovery, this was preceded by the approval of fasudil (a ROCK antagonist) in Japan in 1995 for the treatment of cerebral vasospasm. There are 21 small molecule protein kinase inhibitors that are approved in China, Japan, Europe, and South Korea that are not approved in the United Sates and 75 FDA-approved inhibitors in the United States. Of the 21 agents, eleven target receptor protein-tyrosine kinases, eight inhibit nonreceptor protein-tyrosine kinases, and two block protein-serine/threonine kinases. All 21 drugs are orally bioavailable or topically effective. Of the non-FDA approved drugs, sixteen are prescribed for the treatment of neoplastic diseases, three are directed toward inflammatory disorders, one is used for glaucoma, and fasudil is used in the management of vasospasm. The leading targets of kinase inhibitors approved by both international regulatory agencies and by the FDA are members of the EGFR family, the VEGFR family, and the JAK family. One-third of the 21 internationally approved drugs are not compliant with Lipinski's rule of five for orally bioavailable drugs. The rule of five relies on four parameters including molecular weight, number of hydrogen bond donors and acceptors, and the Log of the partition coefficient.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791-8717, United States.
| |
Collapse
|
29
|
Deng L, Wan L, Liao T, Wang L, Wang J, Wu X, Shi J. Recent progress on tyrosine kinase 2 JH2 inhibitors. Int Immunopharmacol 2023; 121:110434. [PMID: 37315371 DOI: 10.1016/j.intimp.2023.110434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
Tyrosine kinase 2 (TYK2) is a member of the Janus kinase (JAK) family, which can regulate the signaling of multiple pro-inflammatory cytokines, including IL12, IL23 and type I interferon (IFNα/β), and its inhibitors can treat autoimmune diseases caused by the abnormal expression of IL12 and IL23. Interest in TYK2 JH2 inhibitors has increased as a result of safety concerns with JAK inhibitors. This overview introduces TYK2 JH2 inhibitors that are already on the market, including Deucravactinib (BMS-986165), as well as those currently in clinical trials, such as BMS-986202, NDI-034858, and ESK-001.
Collapse
Affiliation(s)
- Lidan Deng
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 610041, China
| | - Li Wan
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 610041, China
| | - Tingting Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| | - Lin Wang
- College of Food and Bioengineering, Xihua University, Chengdu 610039, China
| | - Jie Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou 550002, China
| | - Xianbo Wu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, Sichuan 610041, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
30
|
Xi YJ, Wen R, Zhang R, Dong QR, Zhang HY, Su QY, Zhang SX. Causal association between JAK2 and erectile dysfunction: a Mendelian randomization study. Basic Clin Androl 2023; 33:18. [PMID: 37407943 PMCID: PMC10324256 DOI: 10.1186/s12610-023-00192-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 04/05/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND As one of the most critical proteins in the JAK/STAT signaling pathway, Janus kinase 2 (JAK2) is involved in many biological processes and diseases. Several observational studies have reported the role of JAK2 in erectile dysfunction. However, the causal relationship between JAK2 and erectile dysfunction remains unclear. Here we investigated the causal relationship between JAK2 and erectile dysfunction. RESULTS Genetically predicted JAK2 was causally associated with erectile dysfunction in inverse variance weighting (OR = 1.109, 95% CI = 1.029-1.196, p = 0.007) and weighted median method (OR = 1.117, 95% CI = 1.003-1.245, p = 0.044). No heterogeneity was observed in Cochran Q-test (p = 0.855) and MR-PRESSO (p = 0.866). Pleiotropy was not observed in our study (p = 0.617). CONCLUSIONS These findings highlighted JAK2 as a risk factor for erectile dysfunction and proved the causal relationship between JAK2 and erectile dysfunction, suggesting that targeting JAK2 signaling might be a novel and promising therapeutic candidate in the treatment of erectile dysfunction.
Collapse
Affiliation(s)
- Yu-Jia Xi
- Department of Urology, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, P.R. China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China
| | - Rui Wen
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China
| | - Ran Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China
| | - Qi-Rui Dong
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China
| | - He-Yi Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China
| | - Qin-Yi Su
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, P.R. China
| | - Sheng-Xiao Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Shanxi Province, Taiyuan, P.R. China.
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Shanxi Province, Taiyuan, P.R. China.
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Shanxi Medical University, 382 Wuyi Road, Taiyuan, Shanxi, 030001, P.R. China.
| |
Collapse
|
31
|
Xue C, Yao Q, Gu X, Shi Q, Yuan X, Chu Q, Bao Z, Lu J, Li L. Evolving cognition of the JAK-STAT signaling pathway: autoimmune disorders and cancer. Signal Transduct Target Ther 2023; 8:204. [PMID: 37208335 DOI: 10.1038/s41392-023-01468-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (JAK-STAT) pathway is an evolutionarily conserved mechanism of transmembrane signal transduction that enables cells to communicate with the exterior environment. Various cytokines, interferons, growth factors, and other specific molecules activate JAK-STAT signaling to drive a series of physiological and pathological processes, including proliferation, metabolism, immune response, inflammation, and malignancy. Dysregulated JAK-STAT signaling and related genetic mutations are strongly associated with immune activation and cancer progression. Insights into the structures and functions of the JAK-STAT pathway have led to the development and approval of diverse drugs for the clinical treatment of diseases. Currently, drugs have been developed to mainly target the JAK-STAT pathway and are commonly divided into three subtypes: cytokine or receptor antibodies, JAK inhibitors, and STAT inhibitors. And novel agents also continue to be developed and tested in preclinical and clinical studies. The effectiveness and safety of each kind of drug also warrant further scientific trials before put into being clinical applications. Here, we review the current understanding of the fundamental composition and function of the JAK-STAT signaling pathway. We also discuss advancements in the understanding of JAK-STAT-related pathogenic mechanisms; targeted JAK-STAT therapies for various diseases, especially immune disorders, and cancers; newly developed JAK inhibitors; and current challenges and directions in the field.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qinfan Yao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
32
|
He N, Li L, Li R, Zhang SQ, Wu LH, Guan X, Zhang QY, Jiang T, Yang JB. A Novel Ageladine A Derivative Acts as a STAT3 Inhibitor and Exhibits Potential Antitumor Effects. Int J Mol Sci 2023; 24:ijms24108859. [PMID: 37240202 DOI: 10.3390/ijms24108859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/21/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
The Janus kinase/signal transducer and activator of the transcription 3 (JAK/STAT3) signaling pathway controls multiple biological processes, including cell survival, proliferation, and differentiation. Abnormally activated STAT3 signaling promotes tumor cell growth, proliferation, and survival, as well as tumor invasion, angiogenesis, and immunosuppression. Hence, JAK/STAT3 signaling has been considered a promising target for antitumor therapy. In this study, a number of ageladine A derivative compounds were synthesized. The most effective of these was found to be compound 25. Our results indicated that compound 25 had the greatest inhibitory effect on the STAT3 luciferase gene reporter. Molecular docking results showed that compound 25 could dock into the STAT3 SH2 structural domain. Western blot assays demonstrated that compound 25 selectively inhibited the phosphorylation of STAT3 on the Tyr705 residue, thereby reducing STAT3 downstream gene expression without affecting the expression of the upstream proteins, p-STAT1 and p-STAT5. Compound 25 also suppressed the proliferation and migration of A549 and DU145 cells. Finally, in vivo research revealed that 10 mg/kg of compound 25 effectively inhibited the growth of A549 xenograft tumors with persistent STAT3 activation without causing significant weight loss. These results clearly indicate that compound 25 could be a potential antitumor agent by inhibiting STAT3 activation.
Collapse
Affiliation(s)
- Na He
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Li Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao 266237, China
| | - Rui Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Si-Qi Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Li-Hong Wu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Xian Guan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao 266237, China
| | - Qian-Yue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao 266237, China
| | - Jin-Bo Yang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| |
Collapse
|
33
|
Grant AH, Rodriguez AC, Rodriguez Moncivais OJ, Sun S, Li L, Mohl JE, Leung MY, Kirken RA, Rodriguez G. JAK1 Pseudokinase V666G Mutant Dominantly Impairs JAK3 Phosphorylation and IL-2 Signaling. Int J Mol Sci 2023; 24:ijms24076805. [PMID: 37047778 PMCID: PMC10095075 DOI: 10.3390/ijms24076805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Overactive Janus kinases (JAKs) are known to drive leukemia, making them well-suited targets for treatment. We sought to identify new JAK-activating mutations and instead found a JAK1-inactivating pseudokinase mutation, V666G. In contrast to other pseudokinase mutations that canonically lead to an active kinase, the JAK1 V666G mutation led to under-activation seen by reduced phosphorylation. To understand the functional role of JAK1 V666G in modifying kinase activity we investigated its influence on other JAK kinases and within the Interleukin-2 pathway. JAK1 V666G not only inhibited its own activity, but its presence could inhibit other JAK kinases. These findings provide new insights into the potential of JAK1 pseudokinase to modulate its own activity, as well as of other JAK kinases. Thus, the features of the JAK1 V666 region in modifying JAK kinases can be exploited to allosterically inhibit overactive JAKs.
Collapse
Affiliation(s)
- Alice H. Grant
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Alejandro C. Rodriguez
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Omar J. Rodriguez Moncivais
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Shengjie Sun
- Department of Physics, The University of Texas at El Paso, El Paso, TX 79968, USA
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Lin Li
- Department of Physics, The University of Texas at El Paso, El Paso, TX 79968, USA
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Jonathon E. Mohl
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Mathematical Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Ming-Ying Leung
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
- Computational Science Program, The University of Texas at El Paso, El Paso, TX 79968, USA
- Department of Mathematical Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Robert A. Kirken
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Georgialina Rodriguez
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
34
|
Kadagothy H, Nene S, Amulya E, Vambhurkar G, Rajalakshmi AN, Khatri DK, Singh SB, Srivastava S. Perspective insights of small molecules, phytoconstituents and biologics in the management of psoriasis: A focus on targeting major inflammatory cytokine pathways. Eur J Pharmacol 2023; 947:175668. [PMID: 36958476 DOI: 10.1016/j.ejphar.2023.175668] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023]
Abstract
Psoriasis is an enduring, pruritic and papulosquamous skin ailment that poses a significant burden on public health. It is mainly characterized by hyperkeratosis, acanthosis, parakeratosis, scaly and erythematous plaques. Biomarkers like interleukin-17, interleukin-12 and -23 and tumor necrosis factor-α serve as key drivers of psoriatic pathogenesis. Triggered release of pro-inflammatory cytokines from various up-regulated pathways leads to psoriatic inflammation. Several target moieties like biologics, small molecules and herbal moieties play a fundamental role in the repression of pathogenesis of psoriasis. Biologics and small molecules engaged in the management of psoriasis have been emphasized in detail. An insight into nano-carrier interventions on herbal moieties and clinical aspects of psoriasis are also highlighted. This review emphasizes various pathological targets involved in psoriasis.
Collapse
Affiliation(s)
- Husna Kadagothy
- Department of Pharmaceutics, Mother Theresa Post Graduate and Research Institute of Health Sciences, Puducherry, India
| | - Shweta Nene
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Etikala Amulya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - A N Rajalakshmi
- Department of Pharmaceutics, Mother Theresa Post Graduate and Research Institute of Health Sciences, Puducherry, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
35
|
Liu F, Wang B, Liu Y, Shi W, Hu Z, Chang X, Tang X, Zhang Y, Xu H, He Y. Design, synthesis and biological evaluation of novel N-(methyl-d 3) pyridazine-3-carboxamide derivatives as TYK2 inhibitors. Bioorg Med Chem Lett 2023; 86:129235. [PMID: 36907336 DOI: 10.1016/j.bmcl.2023.129235] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
As a mediator of pro-inflammatory cytokines, TYK2 is an attractive target to treat autoimmunity diseases. Herein, we reported the design, synthesis, and structure-activity relationships (SARs) of N-(methyl-d3) pyridazine-3-carboxamide derivatives as TYK2 inhibitors. Among them, compound 24 exhibited acceptable inhibition activity against STAT3 phosphorylation. Furthermore, 24 showed satisfactory selectivities toward other members of JAK family and performed a good stability profile in liver microsomal assay. Pharmacokinetics (PK) study indicated that compound 24 has reasonable PK exposures. In anti-CD40-induced colitis models, compound 24 was orally highly effective with no significant hERG and CYP isozymes inhibition. These results indicated that compound 24 was worthy of further investigation for the development of anti-autoimmunity diseases agents.
Collapse
Affiliation(s)
- Fei Liu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China; R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Bin Wang
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Yanlong Liu
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Wei Shi
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Zhongyuan Hu
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Xiayun Chang
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Xujing Tang
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Ying Zhang
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Hongjiang Xu
- R&D Institute, Chia Tai Tianqing Pharmaceutical Group Co., Ltd, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Ying He
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, China.
| |
Collapse
|
36
|
Roskoski R. Deucravacitinib is an allosteric TYK2 protein kinase inhibitor FDA-approved for the treatment of psoriasis. Pharmacol Res 2023; 189:106642. [PMID: 36754102 DOI: 10.1016/j.phrs.2022.106642] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 12/31/2022] [Indexed: 02/09/2023]
Abstract
Psoriasis is a heterogeneous, inflammatory, autoimmune skin disease that affects up to 2% of the world's population. There are many treatment modalities including topical medicines, ultraviolet light therapy, monoclonal antibodies, and several oral medications. Cytokines play a central role in the pathogenesis of this disorder including TNF-α, (tumor necrosis factor-α) IL-17A (interleukin-17A), IL-17F, IL-22, and IL-23. Cytokine signaling involves transduction mediated by the JAK-STAT pathway. There are four JAKS (JAK1/2/3 and TYK2) and six STATS (signal transducer and activators of transcription). Janus kinases contain an inactive JH2 domain that is aminoterminal to the active JH1 domain. Under basal conditions, the JH2 domain inhibits the activity of the JH1 domain. Deucravacitinib is an orally effective N-trideuteromethyl-pyridazine derivative that targets and stabilizes the TYK2 JH2 domain and thereby blocks TYK2 JH1 activity. Seven other JAK inhibitors, which target the JAK family JH1 domain, are prescribed for the treatment of neoplastic and other inflammatory diseases. The use of deuterium in the trimethylamide decreases the rate of demethylation and slows the production of a metabolite that is active against a variety of targets in addition to TYK2. A second unique aspect in the development of deucravacitinib is the targeting of a pseudokinase domain. Deucravacitinib is rather specific for TYK2 and its toxic effects are much less than those of the other FDA-approved JAK inhibitors. The successful development of deucravacitinib may stimulate the development of additional pseudokinase ligands for the JAK family and for other kinase families as well.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 3754 Brevard Road, Suite 106, Box 19, Horse Shoe, NC 28742-8814, United States.
| |
Collapse
|
37
|
Zhang X, Lin L, Li L, Hu K, Shao R, Zhang L, Tang L, Zhu M, Ma Y, Yang Y. Janus kinase inhibitor Tofacitinib alleviated acute hepatitis induced by lipopolysaccharide/D-galactosamine in mice. Mol Biol Rep 2023; 50:1477-1485. [PMID: 36507969 DOI: 10.1007/s11033-022-08086-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 11/03/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND The Janus kinase (JAK) is a crucial intracellular signaling hub for numerous cytokines, which is extensively involved in the activation of inflammatory cascade and the induction of inflammatory injury. JAK inhibition provides protective effects in several inflammation-based disorders, but the potential effects of JAK inhibitor in inflammation-based acute hepatitis remain to be investigated. METHODS AND RESULTS Acute hepatitis is induced by Lipopolysaccharide/D-galactosamine (LPS/D-Gal) in mice with or without the JAK inhibitor Tofacitinib administration. The degree of liver injury, the production of pro-inflammatory cytokines and induction of hepatocytes apoptosis were determined. The results indicated that treatment with Tofacitinib decreased the levels of aminotransferases, attenuated the histological abnormalities in liver and decreased the plasma levels of TNF-α and IL-6 in LPS/D-Gal-insulted mice. In addition, Tofacitinib suppressed the activation of the caspase cascade, decreased the level of cleaved caspase-3, and reduced the count of TUNEL-positive cells. CONCLUSION Treatment with Tofacitinib alleviated LPS/D-Gal-induced acute hepatitis. JAK maybe become a promising target for the control of inflammation-based liver disorders.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing, PR China
| | - Ling Lin
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing, PR China
| | - Longjiang Li
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing, PR China
| | - Kai Hu
- Department of Histology and Embryology, Basic Medical College, Chongqing Medical University, Chongqing, PR China
| | - Ruyue Shao
- Clinical Medical School, Chongqing Medical and Pharmaceutical College, Chongqing, PR China
- Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing, PR China
| | - Li Zhang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing, PR China
| | - Li Tang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing, PR China
| | - Min Zhu
- Department of Pathology, Karamay Central Hosptial of XinJiang Karamay, Karamay, Xinjiang, PR China
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Yuhua Ma
- Department of Pathology, Karamay Central Hosptial of XinJiang Karamay, Karamay, Xinjiang, PR China.
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - Yongqiang Yang
- Department of Pathophysiology, Basic Medical College, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
38
|
Sanachai K, Mahalapbutr P, Tabtimmai L, Seetaha S, Kaekratoke N, Chamni S, Azam SS, Choowongkomon K, Rungrotmongkol T. In Silico and In Vitro Study of Janus Kinases Inhibitors from Naphthoquinones. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020597. [PMID: 36677654 PMCID: PMC9866339 DOI: 10.3390/molecules28020597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
Janus kinases (JAKs) are involved in numerous cellular signaling processes related to immune cell functions. JAK2 and JAK3 are associated with the pathogenesis of leukemia and common lymphoid-derived illnesses. JAK2/3 inhibitors could reduce the risk of various diseases by targeting this pathway. Herein, the naphthoquinones were experimentally and theoretically investigated to identify novel JAK2/3 inhibitors. Napabucasin and 2'-methyl napabucasin exhibited potent cell growth inhibition in TF1 (IC50 = 9.57 and 18.10 μM) and HEL (IC50 = 3.31 and 6.65 μM) erythroleukemia cell lines, and they significantly inhibited JAK2/3 kinase activity (in a nanomolar range) better than the known JAK inhibitor, tofacitinib. Flow cytometric analysis revealed that these two compounds induced apoptosis in TF1 cells in a time and dose-dependent manner. From the molecular dynamics study, both compounds formed hydrogen bonds with Y931 and L932 residues and hydrophobically contacted with the conserved hinge region, G loop, and catalytic loop of the JAK2. Our obtained results suggested that napabucasin and its methylated analog were potential candidates for further development of novel anticancer drug targeting JAKs.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panupong Mahalapbutr
- Department of Biochemistry, Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology of North Bangkok, Bangkok 10800, Thailand
| | - Supaphorn Seetaha
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Nantawat Kaekratoke
- Department of Materials Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Supakarn Chamni
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Natural products and Nanoparticles Research Unit (NP2), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Correspondence: (K.C.); (T.R.); Tel.: +66-2-218-5426 (T.R.)
| | - Thanyada Rungrotmongkol
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (K.C.); (T.R.); Tel.: +66-2-218-5426 (T.R.)
| |
Collapse
|
39
|
Properties of FDA-approved small molecule protein kinase inhibitors: A 2023 update. Pharmacol Res 2023; 187:106552. [PMID: 36403719 DOI: 10.1016/j.phrs.2022.106552] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
Owing to the dysregulation of protein kinase activity in many diseases including cancer, this enzyme family has become one of the most important drug targets in the 21st century. There are 72 FDA-approved therapeutic agents that target about two dozen different protein kinases and three of these drugs were approved in 2022. Of the approved drugs, twelve target protein-serine/threonine protein kinases, four are directed against dual specificity protein kinases (MEK1/2), sixteen block nonreceptor protein-tyrosine kinases, and 40 target receptor protein-tyrosine kinases. The data indicate that 62 of these drugs are prescribed for the treatment of neoplasms (57 against solid tumors including breast, lung, and colon, ten against nonsolid tumors such as leukemia, and four against both solid and nonsolid tumors: acalabrutinib, ibrutinib, imatinib, and midostaurin). Four drugs (abrocitinib, baricitinib, tofacitinib, upadacitinib) are used for the treatment of inflammatory diseases (atopic dermatitis, psoriatic arthritis, rheumatoid arthritis, Crohn disease, and ulcerative colitis). Of the 72 approved drugs, eighteen are used in the treatment of multiple diseases. The following three drugs received FDA approval in 2022 for the treatment of these specified diseases: abrocitinib (atopic dermatitis), futibatinib (cholangiocarcinomas), pacritinib (myelofibrosis). All of the FDA-approved drugs are orally effective with the exception of netarsudil, temsirolimus, and trilaciclib. This review summarizes the physicochemical properties of all 72 FDA-approved small molecule protein kinase inhibitors including lipophilic efficiency and ligand efficiency.
Collapse
|
40
|
Krajewski PK, Szepietowski JC. Ruxolitinib cream for the short-term treatment of mild-moderate atopic dermatitis. Expert Rev Clin Immunol 2022; 19:349-356. [PMID: 36542765 DOI: 10.1080/1744666x.2023.2161511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Atopic dermatitis (AD) is a chronic, recurrent, itchy inflammatory skin disease with a vast influence on a patient's quality of life (QoL). Mild-moderate AD was classically managed with the use of topical corticosteroid (TCS) and calcineurin inhibitors (TCI). It was proven that the JAK pathway plays an important role in the development of AD. The introduction of topical JAK inhibitors may revolutionize the classical approach to the management of mild-to-moderate atopic dermatitis. AREAS COVERED This review discussed the role of the JAK pathway in the development and exacerbations of AD with an emphasis on the newly introduced, topical selective JAK1 and JAK2 inhibitor - Ruxolitinib (RUX) cream. It provides an extensive review of pharmacokinetics and pharmacodynamics, efficacy, and safety of RUX cream in clinical trials. EXPERT OPINION Results from phase II and two phase III clinical trials have shown that RUX cream could be a promising topical treatment of mild-moderate AD. Its favorable safety profile and good efficacy make RUX cream a beneficial modality for patients with chronic TCSs and TCIs use. Future studies on younger patients and with a longer observational period are necessary to adequately assess the efficacy and safety of RUX cream in the whole AD population.
Collapse
Affiliation(s)
- Piotr K Krajewski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego Street 1, Wrocław 50-368, Poland
| | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Chalubinskiego Street 1, Wrocław 50-368, Poland
| |
Collapse
|
41
|
Zheng Y, Li X, Kuang L, Wang Y. New insights into the characteristics of DRAK2 and its role in apoptosis: From molecular mechanisms to clinically applied potential. Front Pharmacol 2022; 13:1014508. [PMID: 36386181 PMCID: PMC9649744 DOI: 10.3389/fphar.2022.1014508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 11/27/2022] Open
Abstract
As a member of the death-associated protein kinase (DAPK) family, DAP kinase-associated apoptosis-inducing kinase 2 (DRAK2) performs apoptosis-related functions. Compelling evidence suggests that DRAK2 is involved in regulating the activation of T lymphocytes as well as pancreatic β-cell apoptosis in type I diabetes. In addition, DRAK2 has been shown to be involved in the development of related tumor and non-tumor diseases through a variety of mechanisms, including exacerbation of alcoholic fatty liver disease (NAFLD) through SRSF6-associated RNA selective splicing mechanism, regulation of chronic lymphocytic leukemia and acute myeloid leukemia, and progression of colorectal cancer. This review focuses on the structure, function, and upstream pathways of DRAK2 and discusses the potential and challenges associated with the clinical application of DRAK2-based small-molecule inhibitors, with the aim of advancing DRAK2 research.
Collapse
Affiliation(s)
| | | | | | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
42
|
Ruffolo G, Alfano V, Romagnolo A, Zimmer T, Mills JD, Cifelli P, Gaeta A, Morano A, Anink J, Mühlebner A, Vezzani A, Aronica E, Palma E. GABA A receptor function is enhanced by Interleukin-10 in human epileptogenic gangliogliomas and its effect is counteracted by Interleukin-1β. Sci Rep 2022; 12:17956. [PMID: 36289354 PMCID: PMC9605959 DOI: 10.1038/s41598-022-22806-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/19/2022] [Indexed: 01/24/2023] Open
Abstract
Gangliogliomas (GGs) are low-grade brain tumours that cause intractable focal epilepsy in children and adults. In GG, as in epileptogenic focal malformations (i.e., tuberous sclerosis complex, TSC), there is evidence of sustained neuroinflammation with involvement of the pro-inflammatory cytokine IL-1β. On the other hand, anti-inflammatory mediators are less studied but bear relevance for understanding seizure mechanisms. Therefore, we investigated the effect of the key anti-inflammatory cytokine IL-10 on GABAergic neurotransmission in GG. We assessed the IL-10 dependent signaling by transcriptomic analysis, immunohistochemistry and performed voltage-clamp recordings on Xenopus oocytes microtransplanted with cell membranes from brain specimens, to overcome the limited availability of acute GG slices. We report that IL-10-related mRNAs were up-regulated in GG and slightly in TSC. Moreover, we found IL-10 receptors are expressed by neurons and astroglia. Furthermore, GABA currents were potentiated significantly by IL-10 in GG. This effect was time and dose-dependent and inhibited by blockade of IL-10 signaling. Notably, in the same tissue, IL-1β reduced GABA current amplitude and prevented the IL-10 effect. These results suggest that in epileptogenic tissue, pro-inflammatory mechanisms of hyperexcitability prevail over key anti-inflammatory pathways enhancing GABAergic inhibition. Hence, boosting the effects of specific anti-inflammatory molecules could resolve inflammation and reduce intractable seizures.
Collapse
Affiliation(s)
- Gabriele Ruffolo
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Veronica Alfano
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| | - Alessia Romagnolo
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Till Zimmer
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - James D. Mills
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.83440.3b0000000121901201Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, UK ,grid.452379.e0000 0004 0386 7187Chalfont Centre for Epilepsy, Chalfont St Peter, UK
| | - Pierangelo Cifelli
- grid.158820.60000 0004 1757 2611Department of Applied Clinical and Biotechnological Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandro Gaeta
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
| | - Alessandra Morano
- grid.7841.aDepartment of Human Neuroscience, University of Rome Sapienza, Rome, Italy
| | - Jasper Anink
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.7692.a0000000090126352Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annamaria Vezzani
- grid.4527.40000000106678902Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Eleonora Aronica
- grid.484519.5Department of (Neuro)Pathology, Amsterdam UMC Location University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, The Netherlands ,grid.419298.f0000 0004 0631 9143Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Eleonora Palma
- grid.7841.aDepartment of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy ,grid.18887.3e0000000417581884IRCCS San Raffaele Roma, Rome, Italy
| |
Collapse
|
43
|
Li X, Jiang W, Dong S, Li W, Zhu W, Zhou W. STAT3 Inhibitors: A Novel Insight for Anticancer Therapy of Pancreatic Cancer. Biomolecules 2022; 12:1450. [PMID: 36291659 PMCID: PMC9599947 DOI: 10.3390/biom12101450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/10/2022] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
The signal transducer and activator of transcription (STAT) is a family of intracellular cytoplasmic transcription factors involved in many biological functions in mammalian signal transduction. Among them, STAT3 is involved in cell proliferation, differentiation, apoptosis, and inflammatory responses. Despite the advances in the treatment of pancreatic cancer in the past decade, the prognosis for patients with pancreatic cancer remains poor. STAT3 has been shown to play a pro-cancer role in a variety of cancers, and inhibitors of STAT3 are used in pre-clinical and clinical studies. We reviewed the relationship between STAT3 and pancreatic cancer and the latest results on the use of STAT3 inhibitors in pancreatic cancer, with the aim of providing insights and ideas around STAT3 inhibitors for a new generation of chemotherapeutic modalities for pancreatic cancer.
Collapse
Affiliation(s)
- Xin Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wenkai Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Shi Dong
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wancheng Li
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Weixiong Zhu
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
44
|
Mannucci A, D'Amico F, El Saadi A, Peyrin-Biroulet L, Danese S. Filgotinib for moderately to severely active ulcerative colitis. Expert Rev Gastroenterol Hepatol 2022; 16:927-940. [PMID: 36278878 DOI: 10.1080/17474124.2022.2138857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Filgotinib is an oral Janus kinase type 1 (JAK1) selective inhibitor with demonstrated efficacy and safety in ulcerative colitis (UC). The aim of this review is to summarize the available evidence on pharmacological characteristics, efficacy, and safety of filgotinib in UC. AREAS COVERED Pubmed, Scopus, and Embase databases were searched for all relevant studies reporting the efficacy and safety of filgotinib in patients with moderate to severe UC. We particularly focused on the risk of zoster infection and venous thromboembolism compared to other JAK inhibitors. EXPERT OPINION Filgotinib has remarkable efficacy, safety, and tolerability profiles in the treatment of moderate-to-severe active UC. It can be used in both biologic-naïve and biologic-experienced patients. The rapid mechanism of action and its oral administration route make it a reliable therapeutic option.
Collapse
Affiliation(s)
- Alessandro Mannucci
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Ferdinando D'Amico
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, University of Lorraine, CHRU-Nancy, F-54000, Nancy, France.,Department of Gastroenterology, University of Lorraine, Inserm, NGERE, F-54000, Nancy, France
| | - Silvio Danese
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
45
|
Haikun W, Na W, Dan S. Efficacy and safety of different doses of baricitinib for rheumatoid arthritis: A Bayesian network meta-analysis. Medicine (Baltimore) 2022; 101:e30676. [PMID: 36197174 PMCID: PMC9509125 DOI: 10.1097/md.0000000000030676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND To evaluate the comparative efficacy and safety of baricitinib with different dosages in patients with rheumatoid arthritis (RA). METHODS PubMed, Embase, and the Cochrane Library were retrieved by computer to gather randomized controlled trials (RCTs) of baricitinib for RA from their beginning to September 2021. After 2 researchers independently screened the literature and extracted the data, the risk of bias of included RCTs was assessed, and Bayesian network meta-analysis was performed by GeMTC0.14.3 and Stata15.1 software. RESULTS Ten publications reporting 9 RCTs were included, with 4129 patients randomized to receive 1 of the 7 interventions. Seven interventions were baricitinib 1 mg + conventional disease-modifying antirheumatic drugs (cDMARD), baricitinib 2 mg + cDMARD, baricitinib 4 mg + cDMARD, baricitinib 8 mg + cDMARD, baricitinib 4 mg, placebo + cDMARD, and cDMARD. In the efficacy outcomes at 12 weeks, nearly all doses of baricitinib with or without cDMARD were superior to placebo plus cDMARD and baricitinib 8 mg combined with cDMARD might have the best curative effect in most outcomes. In the efficacy outcomes at 24 weeks, all doses of baricitinib with or without cDMARD were superior to placebo plus cDMARD and baricitinib 4 mg monotherapy might have the best curative effect in most outcomes. The intervention with the highest incidence of adverse events (AEs) might be baricitinib 8 mg combined with cDMARD, and the intervention with the highest incidence of infections might be baricitinib 4 mg combined with cDMARD. CONCLUSIONS Baricitinib 8 mg combined with cDMARDs was suitable for short-term control of RA symptoms, and baricitinib 4 mg was more effective for treating RA over a longer period of time. But attention should be paid for the risk of baricitinib at 4 to 8 mg in clinical application due to the high incidence of AEs and infections.
Collapse
Affiliation(s)
- Wang Haikun
- Department of Pharmacy, The Affiliated Bozhou Hospital of Anhui Medical University (Bozhou People’s Hospital), Bozhou, China
- *Correspondence: Wang Haikun, Department of Pharmacy, The Affiliated Bozhou Hospital of Anhui Medical University (Bozhou People’s Hospital), Bozhou 236800, China (e-mail: )
| | - Wu Na
- Department of Pharmacy, The Affiliated Bozhou Hospital of Anhui Medical University (Bozhou People’s Hospital), Bozhou, China
| | - Su Dan
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| |
Collapse
|
46
|
Sanachai K, Mahalapbutr P, Tabtimmai L, Seetaha S, Kittikool T, Yotphan S, Choowongkomon K, Rungrotmongkol T. Discovery of JAK2/3 Inhibitors from Quinoxalinone-Containing Compounds. ACS OMEGA 2022; 7:33587-33598. [PMID: 36157733 PMCID: PMC9494680 DOI: 10.1021/acsomega.2c04769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
Janus kinases (JAKs) are involved in a wide variety of cell signaling associated with T-cell and B-cell mediated diseases. The pathogenesis of common lymphoid-derived diseases and leukemia cancer has been implicated in JAK2 and JAK3. Therefore, to decrease the risk of these diseases, targeting this pathway using JAK2/3 inhibitors could serve as a valuable research tool. Herein, we used a combination of the computational and biological approaches to identify the quinoxalinone-based dual inhibitors of JAK2/3. First, an in-house library of 49 quinoxalinones was screened by molecular docking. Then, the inhibitory activities of 17 screened compounds against both JAKs as well as against two human erythroleukemia cell lines, TF1 and HEL were examined. The obtained results revealed that several quinoxalinones could potentially inhibit JAK2/3, and among them, ST4j showed strong inhibition against JAKs with the IC50 values of 13.00 ± 1.31 nM for JAK2 and 14.86 ± 1.29 nM for JAK3, which are better than ruxolitinib and tofacitinib. In addition, ST4j potentially inhibited TF1 cells (IC50 of 15.53 ± 0.82 μM) and HEL cells (IC50 of 17.90 ± 1.36 μM), similar to both tofacitinib ruxolitinib. Mechanistically, ST4j inhibited JAK2 autophosphorylation and induced cell apoptosis in dose- and time-dependent manners. From molecular dynamics simulations, ST4j was mainly stabilized by van der Waals interactions, and its hydroxyl group could form hydrogen bonds in the hinge region at residues S936 and R938 of JAK2. This research highlights the potential of ST4j to be a novel therapeutic agent for the treatment of lymphoid-derived diseases and leukemia cancer.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, and Center for Translational Medicine, Faculty of
Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Lueacha Tabtimmai
- Department
of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology of North Bangkok, Bangkok 10800, Thailand
| | - Supaphorn Seetaha
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Tanakorn Kittikool
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Sirilata Yotphan
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok 10400, Thailand
| | - Kiattawee Choowongkomon
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok 10900, Thailand
| | - Thanyada Rungrotmongkol
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- Program
in
Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
47
|
Sanachai K, Mahalapbutr P, Hengphasatporn K, Shigeta Y, Seetaha S, Tabtimmai L, Langer T, Wolschann P, Kittikool T, Yotphan S, Choowongkomon K, Rungrotmongkol T. Pharmacophore-Based Virtual Screening and Experimental Validation of Pyrazolone-Derived Inhibitors toward Janus Kinases. ACS OMEGA 2022; 7:33548-33559. [PMID: 36157769 PMCID: PMC9494641 DOI: 10.1021/acsomega.2c04535] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/26/2022] [Indexed: 06/16/2023]
Abstract
Janus kinases (JAKs) are nonreceptor protein tyrosine kinases that play a role in a broad range of cell signaling. JAK2 and JAK3 have been involved in the pathogenesis of common lymphoid-derived diseases and leukemia cancer. Thus, inhibition of both JAK2 and JAK3 can be a potent strategy to reduce the risk of these diseases. In the present study, the pharmacophore models built based on the commercial drug tofacitinib and the JAK2/3 proteins derived from molecular dynamics (MD) trajectories were employed to search for a dual potent JAK2/3 inhibitor by a pharmacophore-based virtual screening of 54 synthesized pyrazolone derivatives from an in-house data set. Twelve selected compounds from the virtual screening procedure were then tested for their inhibitory potency against both JAKs in the kinase assay. The in vitro kinase inhibition experiment indicated that compounds 3h, TK4g, and TK4b can inhibit both JAKs in the low nanomolar range. Among them, the compound TK4g showed the highest protein kinase inhibition with the half-maximal inhibitory concentration (IC50) value of 12.61 nM for JAK2 and 15.80 nM for JAK3. From the MD simulations study, it could be found that the sulfonamide group of TK4g can form hydrogen bonds in the hinge region at residues E930 and L932 of JAK2 and E903 and L905 of JAK3, while van der Waals interaction also plays a dominant role in ligand binding. Altogether, TK4g, found by virtual screening and biological tests, could serve as a novel therapeutical lead candidate.
Collapse
Affiliation(s)
- Kamonpan Sanachai
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok10330, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen40002, Thailand
| | - Kowit Hengphasatporn
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba305-8577, Ibaraki, Japan
| | - Yasuteru Shigeta
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba305-8577, Ibaraki, Japan
| | - Supaphorn Seetaha
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok10900, Thailand
| | - Lueacha Tabtimmai
- Department
of Biotechnology, Faculty of Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok10800, Thailand
| | - Thierry Langer
- Department
of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14, ViennaA-1090, Austria
| | - Peter Wolschann
- Institute
of Theoretical Chemistry, University of
Vienna, Vienna1090, Austria
| | - Tanakorn Kittikool
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok10400, Thailand
| | - Sirilata Yotphan
- Department
of Chemistry and Center of Excellence for Innovation in Chemistry,
Faculty of Science, Mahidol University, Rama VI Road, Bangkok10400, Thailand
| | - Kiattawee Choowongkomon
- Department
of Biochemistry, Faculty of Science, Kasetsart
University, Bangkok10900, Thailand
| | - Thanyada Rungrotmongkol
- Center
of Excellence in Structural and Computational Biology Research Unit,
Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok10330, Thailand
- Program
in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok10330, Thailand
| |
Collapse
|
48
|
Low ZY, Zabidi NZ, Yip AJW, Puniyamurti A, Chow VTK, Lal SK. SARS-CoV-2 Non-Structural Proteins and Their Roles in Host Immune Evasion. Viruses 2022; 14:v14091991. [PMID: 36146796 PMCID: PMC9506350 DOI: 10.3390/v14091991] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 12/02/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has caused an unprecedented global crisis and continues to threaten public health. The etiological agent of this devastating pandemic outbreak is the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). COVID-19 is characterized by delayed immune responses, followed by exaggerated inflammatory responses. It is well-established that the interferon (IFN) and JAK/STAT signaling pathways constitute the first line of defense against viral and bacterial infections. To achieve viral replication, numerous viruses are able to antagonize or hijack these signaling pathways to attain productive infection, including SARS-CoV-2. Multiple studies document the roles of several non-structural proteins (NSPs) of SARS-CoV-2 that facilitate the establishment of viral replication in host cells via immune escape. In this review, we summarize and highlight the functions and characteristics of SARS-CoV-2 NSPs that confer host immune evasion. The molecular mechanisms mediating immune evasion and the related potential therapeutic strategies for controlling the COVID-19 pandemic are also discussed.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Nur Zawanah Zabidi
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Ashley Jia Wen Yip
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Ashwini Puniyamurti
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
| | - Vincent T. K. Chow
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore 117545, Singapore
- Correspondence: (V.T.K.C.); (S.K.L.)
| | - Sunil K. Lal
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya 47500, Malaysia
- Tropical Medicine & Biology Platform, Monash University, Subang Jaya 47500, Malaysia
- Correspondence: (V.T.K.C.); (S.K.L.)
| |
Collapse
|
49
|
Jiang Y, Zhao T, Zhou X, Xiang Y, Gutierrez‐Castrellon P, Ma X. Inflammatory pathways in COVID-19: Mechanism and therapeutic interventions. MedComm (Beijing) 2022; 3:e154. [PMID: 35923762 PMCID: PMC9340488 DOI: 10.1002/mco2.154] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
The 2019 coronavirus disease (COVID-19) pandemic has become a global crisis. In the immunopathogenesis of COVID-19, SARS-CoV-2 infection induces an excessive inflammatory response in patients, causing an inflammatory cytokine storm in severe cases. Cytokine storm leads to acute respiratory distress syndrome, pulmonary and other multiorgan failure, which is an important cause of COVID-19 progression and even death. Among them, activation of inflammatory pathways is a major factor in generating cytokine storms and causing dysregulated immune responses, which is closely related to the severity of viral infection. Therefore, elucidation of the inflammatory signaling pathway of SARS-CoV-2 is important in providing otential therapeutic targets and treatment strategies against COVID-19. Here, we discuss the major inflammatory pathways in the pathogenesis of COVID-19, including induction, function, and downstream signaling, as well as existing and potential interventions targeting these cytokines or related signaling pathways. We believe that a comprehensive understanding of the regulatory pathways of COVID-19 immune dysregulation and inflammation will help develop better clinical therapy strategies to effectively control inflammatory diseases, such as COVID-19.
Collapse
Affiliation(s)
- Yujie Jiang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Xueyan Zhou
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduPR China
| | - Yu Xiang
- Department of BiotherapyState Key Laboratory of Biotherapy Cancer CenterWest China HospitalSichuan UniversityChengduPR China
| | - Pedro Gutierrez‐Castrellon
- Center for Translational Research on Health Science Hospital General Dr. Manuel Gea GonzalezMinistry of HealthMexico CityMexico
| | - Xuelei Ma
- Department of BiotherapyState Key Laboratory of Biotherapy Cancer CenterWest China HospitalSichuan UniversityChengduPR China
| |
Collapse
|
50
|
Khaledi M, Sameni F, Yahyazade S, Radandish M, Owlia P, Bagheri N, Afkhami H, Mahjoor M, Esmaelpour Z, Kohansal M, Aghaei F. COVID-19 and the potential of Janus family kinase (JAK) pathway inhibition: A novel treatment strategy. Front Med (Lausanne) 2022; 9:961027. [PMID: 36111104 PMCID: PMC9469902 DOI: 10.3389/fmed.2022.961027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Recent evidence proposed that the severity of the coronavirus disease 2019 (COVID-19) in patients is a consequence of cytokine storm, characterized by increased IL-1β, IL-6, IL-18, TNF-α, and IFN-γ. Hence, managing the cytokine storm by drugs has been suggested for the treatment of patients with severe COVID-19. Several of the proinflammatory cytokines involved in the pathogenesis of COVID-19 infection recruit a distinct intracellular signaling pathway mediated by JAKs. Consequently, JAK inhibitors, including baricitinib, pacritinib, ruxolitinib, and tofacitinib, may represent an effective therapeutic strategy for controlling the JAK to treat COVID-19. This study indicates the mechanism of cytokine storm and JAK/STAT pathway in COVID-19 as well as the medications used for JAK/STAT inhibitors.
Collapse
Affiliation(s)
- Mansoor Khaledi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Fatemeh Sameni
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Sheida Yahyazade
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parviz Owlia
- Molecular Microbiology Research Center, Faculty of Medicine, Shahed University, Tehran, Iran
- *Correspondence: Parviz Owlia ;
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Nader Bagheri
| | | | - Mohamad Mahjoor
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Esmaelpour
- Reference Laboratory for Bovine Tuberculosis, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Maryam Kohansal
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzad Aghaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|