1
|
Pi X, Pan M, Zhang Q, Wang Z, Qi Z. Molecular characterization, expression, evolutionary analysis and molecular docking of the Janus activated kinase family members of largemouth bass (Micropterus salmoides). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 167:105375. [PMID: 40288622 DOI: 10.1016/j.dci.2025.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/04/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Janus kinases (JAKs) play crucial roles in the immune responses by binding the cytokine receptors. In the present study, five JAKs family members (JAK1, JAK2a, JAK2b, JAK3, and TYK2) were identified in largemouth bass (Micropterus salmoides). The five JAKs members of largemouth bass contained several conserved protein domains, including a FERM domain, an SH2 domain, a pseudokinase (STyrkc) domain, and a tyrosine kinase (Tyrkc) domain. Phylogenetic analysis revealed the JAK2/2a/2b and JAK3 formed a clade and the JAK1/TYK2 formed another separate clade. Realtime qPCR detection showed that all five JAKs genes were constitutively expressed in ten selected tissues, with highly expression in spleen and head kidney (HK). Following Edwardsiella piscicida infection, the five JAKs genes were significantly upregulated in the spleen and primary hepatocytes at different times post infection. Further selection pressure analysis revealed the five JAKs members underwent negative selection pressures during evolution. The FERM and Tyrkc domains of the five JAK members occurred purifying selection, and involved in interaction with STAT3, which confirmed by protein-protein interaction (PPI) network and molecular docking analysis. Also, molecular docking results indicated that the hydrogens bonds and salt bonds had crucial roles in the JAK/STAT3 complexes formation. Our results indicated that JAKs had important roles in the immune response against bacterial in largemouth bass, providing basis for elucidating the mechanism of JAK/STAT signaling pathway in fish.
Collapse
Affiliation(s)
- Xiangyu Pi
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Mingzhu Pan
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Qihuan Zhang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Zisheng Wang
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Zhitao Qi
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China; Jiangsu Key Laboratory for Exploration and Utilization of Marine Wetland Biological Resources, Yancheng Institute of Technology, Yancheng, China.
| |
Collapse
|
2
|
Chen J, Wang J, Yang W, Zhao L, Su J. Activity Regulation and Conformation Response of Janus Kinase 3 Mediated by Phosphorylation: Exploration from Correlation Network Analysis and Markov Model. J Chem Inf Model 2025. [PMID: 40199555 DOI: 10.1021/acs.jcim.5c00096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The activity of the enzyme JAK3 is modulated by tyrosine phosphorylation, yet the underlying molecular details remain not fully understood. In this study, we employed a GaMD trajectory-based Markov model and correlation network analysis (CNA) to investigate the impact of single phosphorylation (SP) at Y980 (pY980) and double phosphorylation (DP) at Y980/Y981 (pY980/pY981) on the conformational dynamics of JAK3 bound by inhibitors IZA and MI1. The Markov model analysis indicated that both SP and DP result in fewer conformational states and significantly influence the conformational dynamics of the P-loop, αC-helix, and loop1-loop3, while maintaining the hinge region's high rigidity. The CNA findings revealed that phosphorylation alters the communication network among different structural regions of JAK3, providing a rational explanation for how phosphorylation affects the conformational dynamics of the distant P-loop and loop1-loop3. Moreover, the conformational changes mediated by SP and DP further affect the interactions between the inhibitors and the hot spots (L828, V836, E903, Y904, L905, and L956) of JAK3. This work offers valuable theoretical insights into the molecular mechanisms that regulate JAK3 activity.
Collapse
Affiliation(s)
- Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Jian Wang
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Wanchun Yang
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Lu Zhao
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Jing Su
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| |
Collapse
|
3
|
Liu R, Shi X, Zeng W, Wang Y, Yan Z, Deng W, Hui J, Xia R, Mo L, Xu J, Liao T, Miao Y. Novel janus kinase 3 inhibitor ritlecitinib suppresses T and B cell responses to prevent acute cardiac allograft rejection in mice. Clin Immunol 2025; 273:110445. [PMID: 39954912 DOI: 10.1016/j.clim.2025.110445] [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: 11/18/2024] [Revised: 01/30/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Acute rejection is the main contributor to early allograft failure. Current immunosuppressive regimens have shortcomings, such as toxicity and minimal inhibitory effects on B cells. Hence, developing novel, effective, and selective anti-acute rejection drugs is crucial. Therefore, this study aimed to investigate the inhibitory effect of ritlecitinib (PF-06651600), a new janus kinase 3 inhibitor, on T and B cells, as well its preventive effects on acute allograft rejection. A murine cardiac transplantation model coupled with cell culture- and immunohistochemistry-based techniques was used. In vitro assays demonstrated that ritlecitinib inhibits naïve CD4+ T cell differentiation into T helper (Th)1 and Th17 cells, reduces relative inflammatory cytokines, and suppresses CD4+ and CD8+ T cell proliferation. Furthermore, ritlecitinib inhibited B cell activation and differentiation and antibody production. In vivo experiments revealed that ritlecitinib significantly prolongs allograft survival, decreases serum donor-specific antibody immunoglobulin G levels, alleviats allograft damage, and reduces C4d deposition and T, B, and plasma cell infiltration in allografts. Moreover, B and plasma cell percentages and counts were significantly decreased in recipient spleen, lymph nodes, bone marrow, and blood. Overall, ritlecitinib prevented acute rejection in cardiac transplantation by inhibiting T and B cells, suggesting its potential as a novel clinical immunosuppressant.
Collapse
Affiliation(s)
- Rumin Liu
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyi Shi
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenli Zeng
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuchen Wang
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ziyan Yan
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenfeng Deng
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jialiang Hui
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Renfei Xia
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liqian Mo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jian Xu
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tao Liao
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yun Miao
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
4
|
Sathish S, Sohn H, Madhavan T. An In Silico Approach to Uncover Selective JAK1 Inhibitors for Breast Cancer from Life Chemicals Database. Appl Biochem Biotechnol 2025; 197:2508-2543. [PMID: 39760987 DOI: 10.1007/s12010-024-05109-9] [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] [Accepted: 11/19/2024] [Indexed: 01/07/2025]
Abstract
JAK1, a key regulator of multiple oncogenic pathways, is a sought-out target, and its expression in immune cells and tumour-infiltrating lymphocytes (TILs) is associated with a favorable prognosis in breast cancer. JAK1 activates IL-6 via ERBB2 receptor tyrosine kinase signalling and promotes metastatic cancer and STAT3 activation in breast cancer cells. Hence, targeting JAK1 in breast cancer is being explored as a potential therapeutic strategy. A comprehensive in silico approach was utilised in this study to identify selective JAK1 inhibitors from the Life chemicals database. First, we utilised an anticancer focussed library and performed molecular docking to screen against JAK1 protein. The top 10 compounds from docking were taken for cross-docking, to assess the selectivity towards JAK1 target. Lipinski's RO5 was checked for eliminating the compounds that violate rules. Toxicity, biological activity and reactivity for the identified best compounds were predicted by Protox-II server, PASS server and cDFT analysis respectively. MD simulations were carried out to examine the stability and dynamic behaviour of the top leads, including the long-term stability of the ligand-receptor complex and any conformational changes. Lastly, the MM/PBSA method was used to determine the binding free energy of the protein-ligand complex. Our in silico approach has yielded a promising set of compounds F2638-0133, F3408-0020 and F5833-7435 with the potential to selectively target JAK1, a critical player in breast cancer progression. The docking, simulation and MM/PBSA results were compared with standard drug abrocitinib. Identified compounds exhibit favorable binding interactions, electronic properties and robust stability profiles compared to standard drug, making them promising leads for further experimental validation.
Collapse
Affiliation(s)
- Sruthy Sathish
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Honglae Sohn
- Department of Chemistry and Department of Carbon Materials, Chosun University, Gwangju, South Korea
| | - Thirumurthy Madhavan
- Computational Biology Lab, Department of Genetic Engineering, School of Bioengineering, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India.
| |
Collapse
|
5
|
Qiu G, Yu L, Jia L, Cai Y, Chen Y, Jin J, Xu L, Zhu J. Identification of novel covalent JAK3 inhibitors through consensus scoring virtual screening: integration of common feature pharmacophore and covalent docking. Mol Divers 2025; 29:1353-1373. [PMID: 39009908 DOI: 10.1007/s11030-024-10918-5] [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: 05/06/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024]
Abstract
Accumulated research strongly indicates that Janus kinase 3 (JAK3) is intricately involved in the initiation and advancement of a diverse range of human diseases, underscoring JAK3 as a promising target for therapeutic intervention. However, JAK3 shows significant homology with other JAK family isoforms, posing substantial challenges in the development of JAK3 inhibitors. To address these limitations, one strategy is to design selective covalent JAK3 inhibitors. Therefore, this study introduces a virtual screening approach that combines common feature pharmacophore modeling, covalent docking, and consensus scoring to identify novel inhibitors for JAK3. First, common feature pharmacophore models were constructed based on a selection of representative covalent JAK3 inhibitors. The optimal qualitative pharmacophore model proved highly effective in distinguishing active and inactive compounds. Second, 14 crystal structures of the JAK3-covalent inhibitor complex were chosen for the covalent docking studies. Following validation of the screening performance, 5TTU was identified as the most suitable candidate for screening potential JAK3 inhibitors due to its higher predictive accuracy. Finally, a virtual screening protocol based on consensus scoring was conducted, integrating pharmacophore mapping and covalent docking. This approach resulted in the discovery of multiple compounds with notable potential as effective JAK3 inhibitors. We hope that the developed virtual screening strategy will provide valuable guidance in the discovery of novel covalent JAK3 inhibitors.
Collapse
Affiliation(s)
- Genhong Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Li Yu
- School of Inspection and Testing Certification, Changzhou Vocational Institute of Engineering, Changzhou, 213164, Jiangsu, China
| | - Lei Jia
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yanfei Cai
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jian Jin
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China
| | - Jingyu Zhu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| |
Collapse
|
6
|
Rudzki PJ, Jarus-Dziedzic K, Włodarczyk D, Kaza M, Pankiewicz P, Gierczak-Pachulska A, Banach M, Zygmunt B, Piwowarczyk C, Żero P, Rabczenko D, Segiet-Święcicka A, Wieczorek M. First-in-human study of CPL'116 - a dual JAK/ROCK inhibitor - in healthy subjects. Front Pharmacol 2025; 16:1583723. [PMID: 40235539 PMCID: PMC11996792 DOI: 10.3389/fphar.2025.1583723] [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: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
Background CPL'116 is a novel Janus kinase (JAK) and Rho-associated coiled-coil containing protein kinase (ROCK) dual inhibitor and a promising drug candidate for the treatment of inflammatory and fibrotic diseases. We conducted this first-in-human, Phase I clinical trial to evaluate the safety, pharmacokinetics (PK), and exploratory pharmacodynamics (PD) of CPL'116 in healthy subjects. Methods Phase I clinical trial in healthy White volunteers was conducted after single (n = 21, 10-300 mg) and multiple (n = 32, 30-240 mg or placebo, 14-day b.i.d.) administrations of CPL'116 including a food effect study (n = 12, 120 mg). The multiple ascending dose part was double-blinded and placebo-controlled. The primary endpoint was safety evaluation, and the secondary endpoint was PK. Exploratory PD was studied by measuring the inhibition of JAK and ROCK in the blood by assessing STAT1, STAT5, and MLC phosphorylation. Results Safety parameters were comparable between the placebo and active treatment groups, with no clinically meaningful variations in the safety parameters between the cohorts. No deaths or serious adverse events (SAEs) were reported. No influence on hematological parameters (neutrophil count, red cell distribution width, and mean corpuscular volume) was observed. Plasma Cmax and AUC increased proportionally in the dosing range of 60-240 mg. Median tmax ranged 2-3 h. Food increased the absorption of CPL'116. Compared to placebo, CPL'116 at 240 mg dose showed a decrease in the phosphorylation of STAT1 (Days 1 and 14, p < 0.05) and STAT5 (Day 14, p < 0.05). A decrease in MLC phosphorylation indicated a potential trend at p < 0.1. Conclusion CPL'116 was safe and well-tolerated by healthy subjects. The PK profile is well suited for twice-daily administration and justifies further clinical development. Exploratory PD studies indicated the ability of CPL'116 to affect the JAK and ROCK pathways in humans, hinting at its potential therapeutic role in diseases benefiting from its dual mode of action. The positive results of this study indicate the possibility of developing a novel class of therapeutics that address both inflammatory and fibrotic processes. Clinical Trial RegistrationMethods clinicaltrials.gov, identifier NCT04670757.
Collapse
Affiliation(s)
| | | | | | - Michał Kaza
- R&D Center, Celon Pharma S.A., Kazuń Nowy, Poland
| | | | | | | | | | | | - Paweł Żero
- R&D Center, Celon Pharma S.A., Kazuń Nowy, Poland
| | | | | | | |
Collapse
|
7
|
Konen FF, Möhn N, Witte T, Schefzyk M, Wiestler M, Lovric S, Hufendiek K, Jendretzky KF, Gingele S, Schwenkenbecher P, Sühs KW, Friese MA, Klotz L, Pul R, Pawlitzki M, Hagin D, Kleinschnitz C, Meuth SG, Skripuletz T. Disease-modifying strategies: Targeting protein kinases in multiple sclerosis and other autoimmune disorders. Autoimmun Rev 2025; 24:103754. [PMID: 39842533 DOI: 10.1016/j.autrev.2025.103754] [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: 04/19/2024] [Revised: 01/04/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
A wide variety of immunomodulatory therapies are already available for the treatment of multiple sclerosis (MS). Through fundamental insights from basic research with a gain of knowledge in the pathological processes underlying MS, the exploration of additional medical compounds within clinical trials has been ignited. Emerging novel medications with innovative mechanisms of action are being introduced. Those mechanisms of action include a broad therapeutic spectrum of substances targeting various protein kinases, some of which could also be used for the treatment of other autoimmune-mediated diseases. The advancement of new compounds could therefore enable a more personalized approach in treating MS, taking into consideration patients' co-existing autoimmune-mediated diseases. In this review, we discuss potential compounds targeting protein kinases, currently under investigation in clinical trials for various autoimmune diseases that could become viable treatment options for MS and comorbid autoimmune conditions in the future.
Collapse
Affiliation(s)
- Franz Felix Konen
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Nora Möhn
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Torsten Witte
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | - Matthias Schefzyk
- Department of Dermatology, Allergology und Venerology, Hannover Medical School, 30625 Hannover, Germany.
| | - Miriam Wiestler
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, 30625 Hannover, Germany.
| | - Svjetlana Lovric
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany.
| | - Karsten Hufendiek
- University Eye Hospital, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Stefan Gingele
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Kurt-Wolfram Sühs
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany.
| | - Luisa Klotz
- Department of Neurology with Institute of Translational Neurology, University Hospital Muenster, 48149, Muenster, Germany.
| | - Refik Pul
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147 Essen, Germany.
| | - Marc Pawlitzki
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - David Hagin
- Allergy and Clinical Immunology Unit, Department of Medicine, Tel-Aviv Sourasky Medical Center and Sackler Faculty of Medicine, University of Tel Aviv, 6 Weizmann St., Tel-Aviv 6423906, Israel.
| | - Christoph Kleinschnitz
- Department of Neurology, University Medicine Essen, Essen, Germany; Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, 45147 Essen, Germany.
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University Dusseldorf, 40225 Dusseldorf, Germany.
| | - Thomas Skripuletz
- Department of Neurology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
8
|
Diao S, Li L, Zhang J, Ji M, Sun L, Shen W, Wu S, Chen Z, Huang C, Li J. Macrophage-derived CCL1 targets CCR8 receptor in hepatic stellate cells to promote liver fibrosis through JAk/STAT pathway. Biochem Pharmacol 2025; 237:116884. [PMID: 40122149 DOI: 10.1016/j.bcp.2025.116884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Liver fibrosis is caused by liver injury resulting from the wound healing response. According to recent research, the primary factor responsible for liver fibrosis is the activation of hepatic stellate cells (HSCs). C-C motif chemokine ligand 1 (CCL1) is one of several chemokine genes clustered on chromosome 17, which is involved in immune regulation and inflammatory processes. However, the role of CCL1 in liver fibrosis has not been reported. We found that CCL1 secreted by macrophages can target and activate the receptor protein C-C motif chemokine receptor 8 (CCR8) of HSCs, accelerating liver fibrosis progression by activating the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signalling pathway. This suggested that the CCL1-mediated regulation of CCR8 is an important event in liver fibrosis progression. In conclusion, this study identified a novel signalling axis, the CCL1/CCR8/JAK/STAT pathway, which regulates the activation and apoptosis of HSCs, thus providing a novel therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Shaoxi Diao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Liangyun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Jintong Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Minglu Ji
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Lijiao Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Wenwen Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China
| | - Zixiang Chen
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | - Cheng Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China.
| | - Jun Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, PR China; Institute for Liver Diseases of Anhui Medical University, PR China.
| |
Collapse
|
9
|
Bang CH, Park CJ, Kim YS. The Expanding Therapeutic Potential of Deucravacitinib Beyond Psoriasis: A Narrative Review. J Clin Med 2025; 14:1745. [PMID: 40095888 PMCID: PMC11900575 DOI: 10.3390/jcm14051745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/19/2025] Open
Abstract
Deucravacitinib is an allosteric, selective tyrosine kinase 2 (TYK2) inhibitor that has demonstrated significant efficacy in the treatment of psoriasis. TYK2, a member of the Janus kinase (JAK) family, plays a critical role in intracellular signaling pathways for pro-inflammatory cytokines. Unlike traditional JAK inhibitors, which target active domains, deucravacitinib selectively binds to the pseudokinase domain of TYK2. This binding induces a conformational change that locks the enzyme in an inactive state, ensuring superior selectivity for TYK2 over JAK 1/2/3. This unique mechanism specifically inhibits key pro-inflammatory cytokines, including IL-12, IL-23, and type I interferons, critical in the pathogenesis of psoriasis and other immune-mediated diseases. As a result, deucravacitinib represents a promising option for targeted therapy in immune-mediated diseases and may reduce adverse events commonly associated with broader immunosuppressive treatments. Furthermore, its oral administration offers a convenient alternative to injectable biologics, potentially improving patient adherence and treatment satisfaction. This review highlights recent studies suggesting that deucravacitinib may also have therapeutic benefits in psoriatic arthritis, palmoplantar pustulosis, systemic lupus erythematosus, Sjogren's disease, and inflammatory bowel disease. Given its expanding therapeutic potential, deucravacitinib may provide a safer and more effective alternative to current therapies, offering a tailored approach to treatment.
Collapse
Affiliation(s)
- Chul-Hwan Bang
- Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chul-Jong Park
- Department of Dermatology, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 14647, Republic of Korea
| | - Yoon-Seob Kim
- Department of Dermatology, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 14647, Republic of Korea
| |
Collapse
|
10
|
Ciccia F, McGonagle D, Thomas R, Marzo-Ortega H, Martin DA, Yndestad A, Volkov M. JAK inhibition and axial spondyloarthritis: new steps on the path to understanding pathophysiology. Front Immunol 2025; 16:1488357. [PMID: 40103808 PMCID: PMC11913702 DOI: 10.3389/fimmu.2025.1488357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that predominantly affects the sacroiliac joints and spine. Tumor necrosis factor (TNF) and interleukin (IL)-17A are key cytokines in disease pathogenesis and are established axSpA treatment targets. Recently, axSpA treatment options have been complemented by Janus kinase inhibitors (JAKi), which inhibit various cytokines without directly impacting TNF or IL-17 signaling. The effect of JAKi on axSpA remains under investigation: besides a JAK2-mediated (and potentially tyrosine kinase 2 [TYK2]-mediated) effect on the IL-23/IL-17 axis, emerging evidence suggests γδ T cells, type 3 innate lymphoid cells, and mucosa-associated invariant T cells, which are dependent on IL-7 and/or IL-15 and thus on JAK1, are strongly inhibited by JAKi used to treat axSpA. This review summarizes potential effects of JAKi on axSpA and shows evidence from pre-clinical/clinical studies. Greater understanding of the mechanisms of action of available treatments may improve knowledge of axSpA and pave the road for future therapies.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento di Medicina di Precisione, Università della Campania L. Vanvitelli, Naples, Italy
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | | | | |
Collapse
|
11
|
Zhang T, Liu M. Cytokines in age-related eye diseases: pathogenesis and potential targets for innovative therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03926-1. [PMID: 40021512 DOI: 10.1007/s00210-025-03926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Age-related eye diseases (AREDs), such as dry eye disease (DED), age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), are significant worldwide health concerns due to their rising prevalence and debilitating effects. Despite substantial research on the pathobiology of AREDs, the impact of immune-related alterations caused by aging is still not well understood. Tissue-resident cells and invading immune cells in the eye control innate responses in the event of damage or infection. However, as cells age, they gradually lose their ability to perform their protective duties and develop abnormal characteristics. Therefore, the disrupted regulation of immune responses in the eyes of older individuals enhances their vulnerability to and the intensity of eye disorders. Cytokines, immune system components, have a role in developing AREDs by contributing to inflammation. This paper examines the deficiencies in the pathogenic and therapeutic aspects of pro-inflammatory cytokines in AREDs that require further investigation in future studies.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China
| | - Ming Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China.
| |
Collapse
|
12
|
Rana SVS. Mechanistic paradigms of immunotoxicity, triggered by nanoparticles - a review. Toxicol Mech Methods 2025; 35:262-278. [PMID: 39585654 DOI: 10.1080/15376516.2024.2431687] [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: 07/22/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Nanoparticles (NPs) possess the ability to penetrate cells and elicit a rapid and targeted immune response, influenced by their distinct physicochemical properties. These particles can engage with both micro and macromolecules, thereby impacting various downstream signaling pathways that may lead to cell death. This review provides a comprehensive overview of the primary mechanisms contributing to the immunotoxicity of both organic and inorganic nanoparticles. The effects of carbon-based nanomaterials (CNMs), including single-walled carbon nanotubes, multi-walled carbon nanotubes, graphene, and metal oxide nanoparticles, on various immune cell types such as macrophages, neutrophils, monocytes, dendritic cells (DCs), antigen-presenting cells (APCs), and RAW 264.7 cells are examined. The immune responses discussed encompass inflammation, oxidative stress, autophagy, and apoptosis. Additionally, the roles of pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α, and IFN-γ, along with JAK/STAT signaling pathways, are highlighted. The interaction of NPs with oxidative stress pathways, including MAPK signaling and Nrf2/ARE signaling, is also explored. Furthermore, the mechanisms by which nanoparticles induce damage to organelles such as lysosomes, the endoplasmic reticulum, exosomes, and Golgi bodies within the immune system are addressed. The review also emphasizes the genotoxic and epigenetic mechanisms associated with the immunotoxicity of NPs. Recent advancements regarding the immunotherapeutic potential of engineered NPs are reported. The roles of autophagy and apoptosis in the immunotoxicity of NPs merit further investigation. In conclusion, understanding how engineered nanoparticles modulate immune responses may facilitate the prevention and treatment of human diseases, including cancer and autoimmune disorders.
Collapse
Affiliation(s)
- S V S Rana
- Department of Toxicology, Chaudhary Charan Singh University, Meerut, India
| |
Collapse
|
13
|
Kaunitz JD. Sometimes Small Is Beautiful: Discovery of the Janus Kinases (JAK) and Signal Transducer and Activator of Transcription (STAT) Pathways and the Initial Development of JAK Inhibitors for IBD Treatment. Dig Dis Sci 2025; 70:890-898. [PMID: 39827247 PMCID: PMC11919997 DOI: 10.1007/s10620-024-08791-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025]
Abstract
The Janus kinase/signal transducer and activator of transfection (JAK/STAT) system is comprised of multiple cell surface receptors, receptor tyrosine kinases, and signal transducers that are key components of numerous systems involved in malignancy, inflammation, immune surveillance and development, cellular proliferation, metabolism, differentiation, apoptosis, and hematologic disorders, all of which when disrupted can produce severe disease. Nevertheless, small molecule inhibitors of the four known JAKs, termed JAKinibs, have found therapeutic indications for a broad category of diseases. In this perspective, I will summarize the development of JAK inhibitors, whose origins were in antiquity, with particular attention to their use in treating patients with inflammatory bowel disease (IBD). This perspective is accompanied by a companion publication addressing how JAKinibs have forever altered the landscape of IBD therapy.
Collapse
Affiliation(s)
- Jonathan D Kaunitz
- Medical Service, Greater Los Angeles VAMC and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90073, USA.
| |
Collapse
|
14
|
Zeng Q, Ma Y, Cai R, Li X, Luo Y, Zheng B, Wang G, Xu X, Wang X, Liu Z. Direct reprogramming of human fibroblasts into hair-inducing dermal papilla cell-like cells by a single small molecule. Biochem Pharmacol 2025; 233:116744. [PMID: 39798934 DOI: 10.1016/j.bcp.2025.116744] [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: 07/05/2024] [Revised: 12/12/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Dermal papilla cells (DPCs) are a crucial subset of mesenchymal cells in the skin responsible for regulating hair follicle development and growth, making them invaluable for cell-based therapies targeting hair loss. However, obtaining sufficient DPCs with potent hair-inducing abilities remains a persistent challenge. In this study, the Food and Drug Administration (FDA)-approved drug library was utilized to screen small molecules capable of reprogramming readily accessible human skin fibroblasts into functional DPCs. In the initial screening, five candidate small molecules were identified from a pool of 1,817 compounds, and the small molecule peficitinib was further identified by the further hair follicle regeneration experiments. Following peficitinib treatment, fibroblasts derived from primary human foreskin and scalp exhibited the capability to induce hair growth and possessed a molecular profile highly similar to that of primary DPCs. We refer to these cells as dermal papilla cell-like cells (DPC-LCs). Furthermore, transcriptome analysis showed that the wingless/integrated (Wnt) signaling pathway and the transforming growth factor β (TGF-β) signaling pathway, both of which play crucial roles in hair follicle morphogenesis, are upregulated and enriched in these DPC-LCs. These functional DPC-LCs offer a promising avenue for obtaining a plentiful supply of hair-inducing cells, thereby advancing the development of therapeutic strategies for hair loss treatment.
Collapse
Affiliation(s)
- Qinglan Zeng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Yihe Ma
- Department of Respiratory and Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen 518020, China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Ruizhao Cai
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510275, China
| | - Xinxin Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China; Center for Child Care and Mental Health, Shenzhen Children's Hospital Affiliated to Shantou University Medical College, Shenzhen 518026, China
| | - Yilin Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Binkai Zheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China
| | - Gaofeng Wang
- Department of Pastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou 510515, China
| | - Xuejuan Xu
- Department of Endocrinology, The First People's Hospital of Foshan, Foshan 528000, China.
| | - Xusheng Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen 518107, China.
| | - Zhongjie Liu
- Department of Anesthesiology, Shenzhen Children's Hospital, Yitian Road 7019, Shenzhen 518000, China.
| |
Collapse
|
15
|
Abohassan M, Shahrani MMA, Alouda SK, Rajagopalan P. Dibenzo [a, c] phenazin-11-yl(phenyl) methanone (SBLJ23), a novel selective inhibitor targeting JAK2 V617F mutation in myeloproliferative neoplasms. Oncol Res 2025; 33:675-685. [PMID: 40109858 PMCID: PMC11915075 DOI: 10.32604/or.2024.056256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/27/2024] [Indexed: 03/22/2025] Open
Abstract
Background The JAK2V617F mutation plays a crucial part in the pathogenesis of myeloproliferative neoplasms (MPN), which includes polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) leading to aberrant proliferation and survival of hematopoietic cells. Alongside the challenges of drug resistance and side effects, identifying novel compounds that selectively target JAK2V617F could provide more effective and safer therapeutic options for patients with MPNs. Materials and Methods We employed computational approaches like high-throughput virtual screening, molecular dynamics simulations (MDS), and binding free energy calculations to identify inhibitors targeting wild and mutant JAK2 kinases. JAK2V617F positive HEL, wild type JAK2 positive TF-1, and non-cancerous Vero cells were used for in vitro validations. Results SBLJ23 emerged as a top candidate inhibitor with specificity for JAK2V617F. Protein-ligand interaction studies and MDS revealed stable interactions and binding of SBLJ23 over the simulation period, with Root Mean Square Deviation (RMSD) indicating consistent binding after 1t15ns. SBLJ23 displayed a half maximal inhibitory concentration (IC50) value of 522.4 nM against the JAK2 enzyme. The compound exhibited inhibition of cell proliferation in HEL and TF-1 cells, with half maximal cell growth inhibitory concentration (GI50) values of 2.51 and 15.87 µM, respectively. Moreover, SBLJ23 induced G2/M cell cycle arrest in HEL cells to facilitate apoptosis in these cell lines. The compound significantly reduced the percentage of phospho JAK2 and phospho STAT3 in HEL cells. Conclusion High binding affinity, stable interaction profile, favorable binding free energy, and in vitro validations claim SBLJ23 as a potential lead compound against JAK2V617F and suggest further development and optimization towards clinical application in managing myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| | - Mesfer Mohammad Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| | - Sarah Khaled Alouda
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| | - Prasanna Rajagopalan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, 61413, Saudi Arabia
| |
Collapse
|
16
|
Peng S, Meng N, Xie X, Zhu B, Wang B. Identification of Subtypes and Diagnostic Markers Related to Necroptosis in Sepsis. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05201-8. [PMID: 40009340 DOI: 10.1007/s12010-025-05201-8] [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: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Sepsis is a serious systemic infection with a high mortality rate. More and more evidence suggested that necroptosis plays a crucial role in the pathogenesis and progression of sepsis. This study aimed to elucidate the biological function and clinical significance of necroptosis in sepsis, and identify new potential biomarkers to improve the diagnosis and treatment of sepsis. Firstly, we identified 40 differentially expressed necroptosis related genes (DENRGs). Subsequently, a protein interaction (PPI) network of 40 DENRGs was constructed. Based on the key NRGs in the PPI network, the LASSO algorithm was used to screen eight diagnostic-related NRGs in sepsis, and a diagnostic model and risk score were constructed. The ROC analysis results indicated that the eight NRGs diagnostic model has good diagnostic performance (AUC = 0.955). There is a significant difference in risks core between normal samples and sepsis patients. The results of immune infiltration analysis showed that eight diagnostic-related NRGs were significantly correlated with multiple immune cells. Given the clinical significance of necroptosis in sepsis, we identified two molecular subtypes of sepsis based on eight NRGs. The necroptosis score of subtype 1 is significantly lower than that of subtype 2, while the immune score of subtype 1 is significantly higher than that of subtype 2. In summary, we developed and validated a diagnostic model and risk score based on eight NRGs, and identified two completely different subtypes associated with sepsis. Our research may provide new insights into the mechanisms of necroptosis in sepsis and the identification of potential biomarkers.
Collapse
Affiliation(s)
- Sen Peng
- Department of Spinal Surgery, Sunshine Union Hospital of Shandong Province, Weifang, China
| | - Ning Meng
- Department of Neurosurgery, Sunshine Union Hospital of Shandong Province, Weifang, Shandong, 216000, P.R. China
| | - Xia Xie
- Department of Obstetrics and Gynecology, Sunshine Union Hospital of Shandong Province, Weifang, China
| | - Bing Zhu
- Department of Neurosurgery, Sunshine Union Hospital of Shandong Province, Weifang, Shandong, 216000, P.R. China
| | - Bing Wang
- Department of Neurosurgery, Sunshine Union Hospital of Shandong Province, Weifang, Shandong, 216000, P.R. China.
- Department of Trauma Surgery, Sunshine Union Hospital of Shandong Province, Weifang, China.
| |
Collapse
|
17
|
Yadav P, Wairkar S. Tofacitinib in focus: Fascinating voyage from conventional formulations to novel delivery systems. Int J Pharm 2025; 671:125253. [PMID: 39842741 DOI: 10.1016/j.ijpharm.2025.125253] [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: 11/28/2024] [Revised: 01/11/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Tofacitinib, a Janus kinase (JAK) inhibitor, has emerged as a primary therapeutic agent for managing autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, dermatitis and ulcerative colitis. By inhibiting the phosphorylation of JAK enzymes, tofacitinib prevents their activation within the JAK-STAT signaling pathway, which is vital for inflammatory responses. However, the tofacitinib delivery presents significant challenges, including pH-dependent solubility, poor permeability and susceptibility to oral degradation. This review provides an in-depth analysis of current and emerging formulations to enhance the delivery and efficiency of tofacitinib. This review highlights the physicochemical, pharmacodynamic and pharmacokinetic properties of tofacitinib. Additionally, it discusses various strategies, including oral modified release formulations, topical/transdermal delivery utilizing lipid-based and polymeric systems, and parenteral delivery systems. Recent advancements in nanotechnology, such as liposomes, micelles, keratinocyte exosomes, proposomes, proglycosomes, transethosomes, squalenyl nanoparticles and lyotropic liquid crystalline nanoparticles, are explored as potential nanocarriers to existing delivery constraints. The development of advanced tofacitinib delivery systems can address the challenges in its delivery and improve therapeutic outcomes and patient compliance, paving the way for enhanced treatment strategies in autoimmune and inflammatory conditions.
Collapse
Affiliation(s)
- Priti Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra 400056, India.
| |
Collapse
|
18
|
Yashima K, Kurumi H, Yamaguchi N, Isomoto H. Progressing advanced therapies for inflammatory bowel disease: Current status including dual biologic therapy and discontinuation of biologics. Expert Rev Gastroenterol Hepatol 2025:1-20. [PMID: 39968880 DOI: 10.1080/17474124.2025.2469832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 02/20/2025]
Abstract
INTRODUCTION Advanced therapies (ADT) that encompass biological agents and small molecules have been approved for the treatment of inflammatory bowel disease (IBD), broadening the spectrum of available treatment options. Nevertheless, a substantial proportion of patients fail to achieve satisfactory responses, necessitating surgical intervention. Treatment objectives have evolved beyond clinical remission, reduction of inflammation, and mucosal healing, shifting focus toward enhancing the quality of life, acknowledging the profound impact of IBD on physical and mental well-being. AREA COVERED This comprehensive review describes the current landscape of ADT for IBD, including dual biologic therapy (DBT), which involves the combination of two biologics or a single biologic with a small-molecule compound, as well as considerations surrounding the discontinuation of biologics. EXPERT OPINION ADT is the standard treatment for moderate to severe IBD, while DBT appears promising for specific subsets of patients, including those with refractory disease or extraintestinal manifestations. However, these approaches may increase the risk of adverse effects, including malignancy. To optimize individualized treatment strategies in patients with refractory IBD, further trials are needed to refine ADT's predictive value, establish DBT's safety and indications, define biologic discontinuation criteria, and evaluate emerging biomarkers, artificial intelligence, and bowel ultrasound in patient management.
Collapse
Affiliation(s)
- Kazuo Yashima
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hiroki Kurumi
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Naoyuki Yamaguchi
- Department of Endoscopy, Nagasaki University Hospital, Nagasaki, Japan
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
19
|
Anno S, Okano T, Mamoto K, Yamada Y, Mandai K, Orita K, Iida T, Tada M, Inui K, Koike T, Nakamura H. Efficacy and safety of Janus kinase inhibitors in patients with difficult-to-treat rheumatoid arthritis. Mod Rheumatol 2025; 35:225-233. [PMID: 39215592 DOI: 10.1093/mr/roae077] [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: 04/05/2024] [Revised: 06/17/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES This study evaluated the effectiveness of Janus kinase inhibitors (JAKi) in patients with difficult-to-treat rheumatoid arthritis (D2T RA). METHODS This study included 220 patients with RA who were treated with JAKi. Sixty-two patients were naïve to biological disease-modifying antirheumatic drugs (bDMARDs)/JAKi (1st group), 57 patients were failure to one bDMARDs/JAKi (2nd group), and 101 patients were failure to ≥ 2 bDMARDs/JAKi. Of these 101 patients, 25 did not meet the D2T RA criteria (non-D2T RA group) and 76 met the D2T RA criteria (D2T RA group). RESULTS : DAS28-ESR was improved in all groups at 24 weeks (1st: P < .01, 2nd: P < .01, non-D2T RA: P = .01, D2TRA: P = .02), and improvement ratio of DAS28-ESR was not different between DT2RA group and 2nd (P = .73) or non-D2T RA group (P = .68). Glucocorticoid use [odds ratios: 8.67; 95% confidence interval (CI): 1.23-60.90; P = .03] and number of past bDMARD/JAKi uses ≥ 3 (odds ratios: 10.55; 95% CI: 1.39-80.30; P = .02) were risk factors for DAS28-ESR ≥ 3.2 at 24 weeks in the D2T RA group. CONCLUSIONS Clinical efficacy of JAKi in D2T RA group did not differ from that in 2nd and non-D2T RA groups. Glucocorticoid use and multiple bDMARD/JAKi failure were poor prognostic factors for D2T RA.
Collapse
Affiliation(s)
- Shohei Anno
- Department of Orthopaedic Surgery, Yodogawa Christian Hospital, Osaka, Japan
| | - Tadashi Okano
- Center for Senile Degenerative Disorders (CSDD), Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Kenji Mamoto
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Yutaro Yamada
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Koji Mandai
- Mikunioka Mandai Orthopedic Clinic, Sakai, Japan
| | - Kazuki Orita
- Department of Orthopaedic Surgery, Yodogawa Christian Hospital, Osaka, Japan
| | - Takahiro Iida
- Department of Orthopaedic Surgery, Koryokai Hospital, Osaka, Japan
- Department of Orthopeadic Surgery, Takahiro Clinic, Nishinomiya, Japan
| | - Masahiro Tada
- Department of Orthopeadic Surgery, Osaka City General Hospital, Osaka, Japan
| | - Kentaro Inui
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
- Department of Orthopaedic Surgery, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Tatsuya Koike
- Search Institute for Bone and Arthritis Disease (SINBAD) Shirahama Hamayu Hospital, Wakayama, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
20
|
Faris A, Ibrahim IM, Alnajjar R, Hadni H, Bhat MA, Yaseen M, Chakraborty S, Alsakhen N, Shamkh IM, Mabood F, M Naglah A, Ullah I, Ziedan N, Elhallaoui M. QSAR-driven screening uncovers and designs novel pyrimidine-4,6-diamine derivatives as potent JAK3 inhibitors. J Biomol Struct Dyn 2025; 43:757-786. [PMID: 38059345 DOI: 10.1080/07391102.2023.2283168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
This study presents a robust and integrated methodology that harnesses a range of computational techniques to facilitate the design and prediction of new inhibitors targeting the JAK3/STAT pathway. This methodology encompasses several strategies, including QSAR analysis, pharmacophore modeling, ADMET prediction, covalent docking, molecular dynamics (MD) simulations, and the calculation of binding free energies (MM/GBSA). An efficacious QSAR model was meticulously crafted through the employment of multiple linear regression (MLR). The initial MLR model underwent further refinement employing an artificial neural network (ANN) methodology aimed at minimizing predictive errors. Notably, both MLR and ANN exhibited commendable performance, showcasing R2 values of 0.89 and 0.95, respectively. The model's precision was assessed via leave-one-out cross-validation (CV) yielding a Q2 value of 0.65, supplemented by rigorous Y-randomization. , The pharmacophore model effectively differentiated between active and inactive drugs, identifying potential JAK3 inhibitors, and demonstrated validity with an ROC value of 0.86. The newly discovered and designed inhibitors exhibited high inhibitory potency, ranging from 6 to 8, as accurately predicted by the QSAR models. Comparative analysis with FDA-approved Tofacitinib revealed that the new compounds exhibited promising ADMET properties and strong covalent docking (CovDock) interactions. The stability of the new discovered and designed inhibitors within the JAK3 binding site was confirmed through 500 ns MD simulations, while MM/GBSA calculations supported their binding affinity. Additionally, a retrosynthetic study was conducted to facilitate the synthesis of these potential JAK3/STAT inhibitors. The overall integrated approach demonstrates the feasibility of designing novel JAK3/STAT inhibitors with robust efficacy and excellent ADMET characteristics that surpass Tofacitinib by a significant margin.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdelmoujoud Faris
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science, University of Benghazi, Benghazi, Libya
| | - Hanine Hadni
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mashooq Ahmad Bhat
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Yaseen
- Institute of Chemical Sciences, University of Swat, Main Campus, Charbagh, Swat, Pakistan
| | - Souvik Chakraborty
- Department of Physiology, Bhairab Ganguly College, Belghoria, Kolkata, West Bengal, India
| | - Nada Alsakhen
- Department of Chemistry, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Israa M Shamkh
- Botany and Microbiology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Fazal Mabood
- Institute of Chemical Sciences, University of Swat, Main Campus, Charbagh, Swat, Pakistan
| | - Ahmed M Naglah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ihsan Ullah
- Institute of Chemical Sciences, University of Swat, Main Campus, Charbagh, Swat, Pakistan
| | - Noha Ziedan
- Department of Physical, Mathematical and Engineering Science, Faculty of Science, Business and Enterprise, University of Chester, Chester, UK
| | - Menana Elhallaoui
- LIMAS, Department of Chemical Sciences, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
21
|
Kaul A, Hope H, Xu C, Basavalingappa R, Binz SK, Boily C, Bradley Z, Burt D, Emanuel C, Fairchild J, Egan S, Hildebrand A, Howell V, Huang H, Huff E, Iken A, Knapik S, Lawrence M, Lin H, Lu JW, Mattingly J, McGraw D, McGraw N, Mnich S, Morton W, Ortmann R, Piccinni-Ash T, Saer R, da Silva CS, Stillwell L, Taylor W, Warner E, Wrightstone A, Jacobsen EJ, Anderson DR, Monahan J. Characterization of the dual ITK/JAK3 small molecule covalent inhibitor ATI-2138. J Pharmacol Exp Ther 2025; 392:100054. [PMID: 40023606 DOI: 10.1016/j.jpet.2024.100054] [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: 07/26/2024] [Accepted: 11/12/2024] [Indexed: 03/04/2025] Open
Abstract
Aclaris Therapeutics Inc (ATI)-2138 is a novel investigational covalent inhibitor of interleukin-2-inducible T cell kinase (ITK), resting lymphocyte kinase, and Janus kinase 3 (JAK3) in development for the treatment of autoimmune and inflammatory diseases. In this study, we evaluated the inhibitory effects of ATI-2138 on ITK and JAK3 signaling in cells and preclinical animal models and assessed the safety, tolerability, pharmacokinetics, and pharmacodynamics of ATI-2138 in healthy human participants. ATI-2138 potently, selectively, and irreversibly inhibited ITK, resting lymphocyte kinase, and JAK3 kinases with similar potency observed against ITK and JAK3 in biochemical and immune cell signaling assays. Translation from cellular and whole blood studies to in vivo models was observed, wherein ATI-2138 demonstrated disease-modifying activity in 2 rodent models of arthritis and an adoptive T cell model of colitis. In single and multiple ascending dose studies in healthy human participants, ATI-2138 had a favorable safety profile with linear pharmacokinetics. Biomarkers linked to both ITK and JAK3 activity were inhibited with ATI-2138 in an exposure-, dose-, and time-dependent manner and correlated with enzyme, cellular, whole blood, and rodent studies, thereby demonstrating predictive translational properties. As a potential first-in-class dual inhibitor of ITK and JAK3, ATI-2138 may be useful in the treatment of immunoinflammatory diseases. SIGNIFICANCE STATEMENT: Aclaris Therapeutics Inc (ATI)-2138 is a novel covalent inhibitor of interleukin-2-inducible T cell kinase (ITK)/resting lymphocyte kinase and Janus kinase 3 (JAK3). ATI-2138 inhibits JAK3 and ITK in enzyme and functional cellular assays, demonstrates disease-modifying activity in rodent models of arthritis and colitis, and inhibits biomarkers linked to both ITK and JAK3 activity in healthy human participants. With this dual kinase activity against components of these inflammatory signaling pathway, ATI-2138 has the potential for enhanced therapeutic efficacy in the treatment of autoimmune and chronic inflammatory disease.
Collapse
Affiliation(s)
- Aparna Kaul
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Heidi Hope
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Canxin Xu
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | - Sara K Binz
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Chad Boily
- Aclaris Therapeutics, Inc, Wayne, Pennsylvania
| | | | - David Burt
- Aclaris Therapeutics, Inc, Wayne, Pennsylvania
| | | | | | - Sarah Egan
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | - Huiyan Huang
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Emma Huff
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Abbygail Iken
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | - Huawen Lin
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | - Dean McGraw
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Nancy McGraw
- Confluence Discovery Technologies, St. Louis, Missouri
| | - Stephen Mnich
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | | | - Rafael Saer
- Confluence Discovery Technologies, St. Louis, Missouri
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang YF, Chen CY, Lei L, Zhang Y. Regulation of the microglial polarization for alleviating neuroinflammation in the pathogenesis and therapeutics of major depressive disorder. Life Sci 2025; 362:123373. [PMID: 39756509 DOI: 10.1016/j.lfs.2025.123373] [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: 06/06/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Major depressive disorder (MDD), as a multimodal neuropsychiatric and neurodegenerative illness with high prevalence and disability rates, has become a burden to world health and the economy that affects millions of individuals worldwide. Neuroinflammation, an atypical immune response occurring in the brain, is currently gaining more attention due to its association with MDD. Microglia, as immune sentinels, have a vital function in regulating neuroinflammatory reactions in the immune system of the central nervous system. From the perspective of steady-state branching states, they can transition phenotypes between two extremes, namely, M1 and M2 phenotypes are pro-inflammatory and anti-inflammatory, respectively. It has an intermediate transition state characterized by different transcriptional features and the release of inflammatory mediators. The timing regulation of inflammatory cytokine release is crucial for damage control and guiding microglia back to a steady state. The dysregulation can lead to exorbitant tissue injury and neuronal mortality, and targeting the cellular signaling pathway that serves as the regulatory basis for microglia is considered an essential pathway for treating MDD. However, the specific intervention targets and mechanisms of microglial activation pathways in neuroinflammation are still unclear. Therefore, the present review summarized and discussed various signaling pathways and effective intervention targets that trigger the activation of microglia from its branching state and emphasizes the mechanism of microglia-mediated neuroinflammation associated with MDD.
Collapse
Affiliation(s)
- Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
23
|
Solano Q, Uttal S, Higgins PDR, Berinstein JA. Disseminated Chickenpox Following Live Varicella Vaccination in a Crohn's Disease Patient on Combination Immunosuppression. Case Rep Gastrointest Med 2025; 2025:6088333. [PMID: 39872708 PMCID: PMC11772060 DOI: 10.1155/crgm/6088333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
Novel therapeutics used in the treatment of inflammatory bowel disease pose an increased risk of viral reactivation in patients. We present a case of a patient with refractory Crohn's disease (CD) who developed primary varicella (chickenpox) of a vaccine-viral strain after receiving combination immunosuppression with high-dose corticosteroids, tumor necrosis factor inhibitor (TNFi), and a Janus kinase inhibitor (JAKi) in the hospital. While this patient recovered and did not experience long term adverse effects, her case provides an opportunity for improvement. To improve safety, healthcare facilities should develop protocols that use electronic medical records enhanced with clinical decision support systems to identify and protect immunocompromised patients from inappropriate live vaccinations.
Collapse
Affiliation(s)
- Quintin Solano
- Department of Internal Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Sarah Uttal
- Department of Gastroenterology and Hepatology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Peter D. R. Higgins
- Department of Gastroenterology and Hepatology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Jeffrey A. Berinstein
- Department of Gastroenterology and Hepatology, Michigan Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Gosia M, Doshi G, Bagwe Parab S, Godad A. Innovative Approaches to Psoriasis: Small Molecules Targeting Key Signaling Pathways. Immunol Invest 2025:1-37. [PMID: 39819440 DOI: 10.1080/08820139.2025.2449960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
BACKGROUND Psoriasis (Pso) is a chronic, immune-mediated dermatological condition characterized by dysregulated inflammatory responses and the hyperproliferation of keratinocytes. Biologics, which target specific cytokines such as IL-17 and IL-23, have revolutionized the management by addressing key drivers of its pathophysiology. Despite their efficacy, biologics are not without limitations, including the need for intermittent administration and ongoing monitoring. In contrast, small molecules offer a promising alternative by selectively inhibiting key signaling pathways that modulate pro-inflammatory cytokines involved in the inflammatory cascade. METHODS AND RESULTS This review suggests a new therapeutic strategy for Pso treatment, emphasizing the intricate relationships between small molecules and important signaling pathways involved in the pathophysiology of skin conditions. Improving treatment outcomes and reducing the side effects associated with conventional medicines, this review aims to better understand how tailored small-molecule inhibitors might efficiently control these pathways. This creative approach promotes the creation of individualized treatment plans that can greatly enhance the quality of life of patients with Psoby utilizing the knowledge gathered from recent developments in signaling pathway research. CONCLUSION This review delves into the molecular mechanisms underlying Pso and explores how small molecules can be harnessed to enhance treatment outcomes, presenting a new paradigm for managing this chronic skin disorder.
Collapse
Affiliation(s)
- Meeral Gosia
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Siddhi Bagwe Parab
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Angel Godad
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
25
|
Yagi S, Fukui H, Ikenouchi M, Shiraishi T, Kaku K, Wakita M, Takagi Y, Sato T, Kawai M, Kamikozuru K, Yokoyama Y, Takagawa T, Tomita T, Shinzaki S. Continuous Treatment with Tofacitinib but Not Filgotinib Is Effective in Non-Responders with Active Ulcerative Colitis: A Propensity Score-Matching Analysis. J Clin Med 2025; 14:217. [PMID: 39797304 PMCID: PMC11721360 DOI: 10.3390/jcm14010217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Few studies have compared the efficacy and safety of Janus kinase (JAK) inhibitors in patients with ulcerative colitis (UC). We compared the real-world effectiveness and safety of tofacitinib (TOF) and filgotinib (FIL) as induction therapy for UC by propensity score-matching analysis. Methods: We enrolled 230 patients with active UC who received either TOF (n = 197) or FIL (n = 33) as induction therapy. The primary outcome was the clinical response at week 8, and the secondary outcomes were the clinical response/remission rates from weeks 2-8, including the course of patients without a clinical response/remission at week 4. Results: Propensity score-matching analysis revealed that the clinical response rate gradually increased to 72.2% at 8 weeks in the TOF group, whereas it tended to decrease to 48.5% in the FIL group. Clinical remission rates increased from 2 (36.7% vs. 36.7%) to 8 weeks (63.6% vs. 48.5%) after treatment in the TOF and FIL groups, respectively. The clinical response rate was higher in the TOF group than in the FIL group at week 8 in patients without a clinical response at week 4 (38.5% vs. 0%; p = 0.011). The clinical remission rate was also higher in the TOF group than in the FIL group at week 8 in patients without clinical remission at week 4 (50.0% vs. 16.7%; p = 0.046). The incident rates of infection and anemia were higher in the TOF group than in the FIL group. Conclusions: TOF may be more effective than FIL at 8 weeks for patients with UC who do not respond to treatment within the first 4 weeks.
Collapse
Affiliation(s)
- Soichi Yagi
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Hirokazu Fukui
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Maiko Ikenouchi
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Tetsuya Shiraishi
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Koji Kaku
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Midori Wakita
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Yasuhiro Takagi
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Toshiyuki Sato
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Mikio Kawai
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Koji Kamikozuru
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Yoko Yokoyama
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Tetsuya Takagawa
- Center for Clinical Research and Education, Hyogo Medical University, Nishinomiya 663-8501, Hyogo, Japan;
| | - Toshihiko Tomita
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| | - Shinichiro Shinzaki
- Department of Gastroenterology, Faculty of Medicine, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan; (S.Y.); (H.F.); (M.I.); (T.S.); (K.K.); (M.W.); (Y.T.); (T.S.); (M.K.); (K.K.); (Y.Y.); (T.T.)
| |
Collapse
|
26
|
Virtanen A, Kettunen V, Musta K, Räkköläinen V, Knapp S, Haikarainen T, Silvennoinen O. Molecular basis of JAK kinase regulation guiding therapeutic approaches: Evaluating the JAK3 pseudokinase domain as a drug target. Adv Biol Regul 2025; 95:101072. [PMID: 39755448 DOI: 10.1016/j.jbior.2024.101072] [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: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025]
Abstract
Janus kinases (JAK1-3, TYK2) are critical mediators of cytokine signaling and their role in hematological and inflammatory and autoimmune diseases has sparked widespread interest in their therapeutic targeting. JAKs have unique tandem kinase structure consisting of an active tyrosine kinase domain adjacent to a pseudokinase domain that is a hotspot for pathogenic mutations. The development of JAK inhibitors has focused on the active kinase domain and the developed drugs have demonstrated good clinical efficacy but due to off-target inhibition cause also side-effects and carry a black box warning limiting their use. Our understanding of the regulatory function of the pseudokinase domain in physiological and pathological signaling has improved substantially. The pseudokinase domain maintains the inactive state of JAKs in the absence of cytokine stimulation but it has also a key role in physiological and mutation-driven activation process. Furthermore, the pseudokinase domain has favourable structural characteristics for selective targeting of cytokine signaling, such as unique mode of ATP-binding, and the first pseudokinase targeting inhibitor for TYK2 has been approved for clinical use. Here we describe the recent functional and structural knowledge of JAK signaling and their therapeutic targeting, and present data evaluating the druggability of the JAK3 pseudokinase domain.
Collapse
Affiliation(s)
- Anniina Virtanen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpönkatu 34, 33014, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, P.O. Box 56, 00014, Finland
| | - Vivian Kettunen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpönkatu 34, 33014, Finland
| | - Kirsikka Musta
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpönkatu 34, 33014, Finland
| | - Veera Räkköläinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpönkatu 34, 33014, Finland
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany; Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Teemu Haikarainen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpönkatu 34, 33014, Finland; Department of Microbiology, Fimlab Laboratories, P.O.Box 66, 33013, Tampere, Finland
| | - Olli Silvennoinen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpönkatu 34, 33014, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, P.O. Box 56, 00014, Finland; Department of Microbiology, Fimlab Laboratories, P.O.Box 66, 33013, Tampere, Finland.
| |
Collapse
|
27
|
Mohanty C, Singh CK, Daccache JA, Damsky W, Kendziorski C, Yan D, Prasad A, Zhang D, Keenan T, Drolet B, Ahmad N, Shields BE. Granuloma Annulare Exhibits Mixed Immune and Macrophage Polarization Profiles with Spatial Transcriptomics. J Invest Dermatol 2025; 145:109-121. [PMID: 38844128 DOI: 10.1016/j.jid.2024.04.024] [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: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 07/16/2024]
Abstract
Granuloma annulare (GA) is an idiopathic condition characterized by granulomatous inflammation in the skin. Prior studies have suggested that GA develops from various triggers, leading to a complex interplay involving innate and adaptive immunity, tissue remodeling, and fibrosis. Macrophages are the major immune cells comprising GA granulomas; however, the molecular drivers and inflammatory signaling cascade behind macrophage activation are poorly understood. Histologically, GA exhibits both palisaded and interstitial patterns on histology; however, the molecular composition of GA at the spatial level remains unexplored. GA is a condition without Food and Drug Administration-approved therapies despite the significant impact of GA on QOL. Spatial transcriptomics is a valuable tool for profiling localized, genome-wide gene expression changes across tissues, with emerging applications in clinical medicine. To improve our understanding of the spatially localized gene expression patterns underlying GA, we profiled the spatial gene expression landscape from 6 patients with GA. Our findings revealed mixed T helper 1 and T helper 2 signals comprising the GA microenvironment and spatially distinct M1 and M2 macrophage polarization characteristics. IFN-γ and TNF signals emerged as important regulators of GA granulomatous inflammation, and IL-32 emerged as a key driver of granulomatous inflammation. Overall, our spatial transcriptomics data indicate that GA exhibits mixed immune and macrophage polarization.
Collapse
Affiliation(s)
- Chitrasen Mohanty
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chandra K Singh
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph A Daccache
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Pathology, NYU Langone Health, New York, New York, USA
| | - William Damsky
- Department of Pathology, NYU Langone Health, New York, New York, USA; Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Di Yan
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aman Prasad
- Department of Dermatology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Donglin Zhang
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tom Keenan
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Beth Drolet
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nihal Ahmad
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Dermatology, William S. Middleton Memorial Veterans' Hospital, Madison, Wisconsin, USA
| | - Bridget E Shields
- Department of Dermatology, The School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
28
|
Xia L, Li H, Long L, Ruan W, Ma J, Xu S, Qiao D. Research progress on the pathogenesis of psoriasis and its small molecule inhibitors. Arch Pharm (Weinheim) 2025; 358:e2400621. [PMID: 39686874 DOI: 10.1002/ardp.202400621] [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: 07/31/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024]
Abstract
Psoriasis is a prevalent chronic systemic immune disease characterized by T-cellmediated hyperproliferation of keratinized cells. Among its various manifestations, plaque-type psoriasis is the most common. Treatment options for psoriasis encompass topical medications, biological therapies, phototherapy techniques, and others. However, traditional treatments are associated with numerous side effects. In contrast, targeted therapy has garnered increasing attention due to its high selectivity, strong safety profile, and favorable therapeutic outcomes. Patients with psoriasis lesions exhibit elevated levels of proinflammatory cytokines compared with the general population. These proinflammatory cytokines have been implicated in mediating psoriasis pathogenesis by inducing keratinocyte proliferation through multiple signaling pathways within the body. This study will delve into the Janus kinase-signal transducers and activators of transcription, phosphatidylinositol 3 kinase (PI3K)-protein kinase B (PKB, also known as AKT), and nuclear factor Kappa-light-chain-enhancer of activated B cells signaling pathways to elucidate their roles in mediating psoriasis pathogenesis. In addition, we will summarize potential targets relevant to the treatment of psoriasis and discuss the design and activity assessment of their inhibitors. It also provides new insights for further in-depth study of psoriasis and development of novel molecularly targeted inhibitors.
Collapse
Affiliation(s)
- Lulu Xia
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Hongxin Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Li Long
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Wei Ruan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Jiajia Ma
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| |
Collapse
|
29
|
Twomey RE, Perper SJ, Westmoreland SV, Srinivasan S, Mathieu SL, Frank KE, Karman J, Long AJ, Housley WJ, Clarke SH. Therapeutic JAK1 Inhibition Reverses Lupus Nephritis in a Mouse Model and Demonstrates Transcriptional Changes Consistent With Human Disease. ACR Open Rheumatol 2024; 6:900-911. [PMID: 39364807 PMCID: PMC11638135 DOI: 10.1002/acr2.11745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVE Janus kinase family members are essential for signaling by multiple cytokines, including many implicated in systemic lupus erythematosus (SLE) pathogenesis. To test whether inhibition of JAK1 signaling can be efficacious in SLE, we used a JAK1-selective inhibitor (ABT-317) and evaluated its ability to ameliorate disease in murine SLE. METHODS Efficacy of ABT-317 was evaluated using NZB/W-F1 mice treated prophylactically and therapeutically. Primary endpoints were proteinuria, survival, and saliva production. Other endpoints included histological analysis of kidneys and salivary glands, flow cytometric analysis of splenic cell populations, and gene expression analysis by RNA sequencing in the kidneys, salivary glands, and blood. Publicly available human kidney gene transcription data were used to assess the translatability of the mouse findings. RESULTS ABT-317 was efficacious when dosed prophylactically and prevented disease for up to two months after treatment cessation. When dosed therapeutically, ABT-317 quickly reversed severe proteinuria and restored saliva production, as well as diminished kidney and salivary gland inflammation. ABT-317-induced changes in glomerular morphology coincided with normalization of a human nephrotic gene signature, suggesting translatability to human lupus nephritis (LN). CONCLUSION JAK1 inhibition prevented and reversed kidney and salivary gland manifestations of murine lupus with long-lasting effects after treatment cessation. These data, along with the presence of JAK1 and nephrotic gene signatures in human LN glomeruli, suggest that a JAK1-selective inhibitor may be an effective therapeutic in the treatment of human SLE and LN.
Collapse
|
30
|
Sian-Hulsmann J, Riederer P. Virus-induced brain pathology and the neuroinflammation-inflammation continuum: the neurochemists view. J Neural Transm (Vienna) 2024; 131:1429-1453. [PMID: 38261034 PMCID: PMC11608394 DOI: 10.1007/s00702-023-02723-5] [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: 09/21/2023] [Accepted: 11/18/2023] [Indexed: 01/24/2024]
Abstract
Fascinatingly, an abundance of recent studies has subscribed to the importance of cytotoxic immune mechanisms that appear to increase the risk/trigger for many progressive neurodegenerative disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis, and multiple sclerosis. Events associated with the neuroinflammatory cascades, such as ageing, immunologic dysfunction, and eventually disruption of the blood-brain barrier and the "cytokine storm", appear to be orchestrated mainly through the activation of microglial cells and communication with the neurons. The inflammatory processes prompt cellular protein dyshomeostasis. Parkinson's and Alzheimer's disease share a common feature marked by characteristic pathological hallmarks of abnormal neuronal protein accumulation. These Lewy bodies contain misfolded α-synuclein aggregates in PD or in the case of AD, they are Aβ deposits and tau-containing neurofibrillary tangles. Subsequently, these abnormal protein aggregates further elicit neurotoxic processes and events which contribute to the onset of neurodegeneration and to its progression including aggravation of neuroinflammation. However, there is a caveat for exclusively linking neuroinflammation with neurodegeneration, since it's highly unlikely that immune dysregulation is the only factor that contributes to the manifestation of many of these neurodegenerative disorders. It is unquestionably a complex interaction with other factors such as genetics, age, and environment. This endorses the "multiple hit hypothesis". Consequently, if the host has a genetic susceptibility coupled to an age-related weakened immune system, this makes them more susceptible to the virus/bacteria-related infection. This may trigger the onset of chronic cytotoxic neuroinflammatory processes leading to protein dyshomeostasis and accumulation, and finally, these events lead to neuronal destruction. Here, we differentiate "neuroinflammation" and "inflammation" with regard to the involvement of the blood-brain barrier, which seems to be intact in the case of neuroinflammation but defect in the case of inflammation. There is a neuroinflammation-inflammation continuum with regard to virus-induced brain affection. Therefore, we propose a staging of this process, which might be further developed by adding blood- and CSF parameters, their stage-dependent composition and stage-dependent severeness grade. If so, this might be suitable to optimise therapeutic strategies to fight brain neuroinflammation in its beginning and avoid inflammation at all.
Collapse
Affiliation(s)
- Jeswinder Sian-Hulsmann
- Department of Human Anatomy and Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| | - Peter Riederer
- University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark, Winslows Vey 18, 5000, Odense, J.B, Denmark.
| |
Collapse
|
31
|
Ouassaf M, Bourougaa L, Bahaz F, Alhatlani BY. Exploring the Antiviral Potential of Artemisia annua Through JAK-STAT Pathway Targeting: A Network Pharmacology Approach. Pharmaceuticals (Basel) 2024; 17:1539. [PMID: 39598448 PMCID: PMC11597232 DOI: 10.3390/ph17111539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Artemisia annua, a plant with antiviral potential, has shown promise against various viral infections, yet its mechanisms of action are not fully understood. This study explores A. annua's antiviral effects using network pharmacology and molecular docking, focusing on key active compounds and their interactions with viral protein targets, particularly within the JAK-STAT signaling pathway-a critical mediator of immune responses to viral infections. METHODS From the TCMSP database, we identified eight active compounds and 335 drug targets for A. annua, with 19 intersecting targets between A. annua compounds and viral proteins. A protein-protein interaction (PPI) network highlighted 10 key hub genes, analyzed further through Gene Ontology (GO) and KEGG pathways to understand their immune and antiviral roles. ADMET properties of the active compound Patuletin (MOL004112) were assessed, followed by 200 ns molecular dynamics simulations to examine its stability in complex with JAK2. RESULTS PPI analysis identified JAK2, MAPK3, MAPK1, JAK1, PTPN1, HSPA8, TYK2, RAF1, MAPT, and HMOX1 as key hub genes, with JAK2 emerging as a critical regulator of immune and antiviral pathways. ADMET analysis confirmed Patuletin's favorable pharmacokinetic properties, and molecular dynamics simulations showed a stable Patuletin-JAK2 complex, with FEL analysis indicating minimal disruption to JAK2's intrinsic flexibility. CONCLUSIONS These findings highlight JAK2 as a promising target in the antiviral activity of A. annua compounds, particularly Patuletin, supporting its potential as an antiviral agent and providing a foundation for further research on A. annua's therapeutic applications.
Collapse
Affiliation(s)
- Mebarka Ouassaf
- Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 07000, Algeria;
| | - Lotfi Bourougaa
- Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 07000, Algeria;
| | - Farial Bahaz
- Laboratory of Organic Materials and Heterochemistry, Echahid Cheikh Larbi Tebessi University, Tebessa 12000, Algeria;
| | - Bader Y. Alhatlani
- Unit of Scientific Research, Applied College, Qassim University, Buraydah 52571, Saudi Arabia
| |
Collapse
|
32
|
Bukowska P, Bralewska M, Pietrucha T, Sakowicz A. Nutraceuticals as Modulators of Molecular Placental Pathways: Their Potential to Prevent and Support the Treatment of Preeclampsia. Int J Mol Sci 2024; 25:12167. [PMID: 39596234 PMCID: PMC11594370 DOI: 10.3390/ijms252212167] [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: 10/31/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Preeclampsia (PE) is a serious condition characterized by new-onset hypertension and proteinuria or organ dysfunction after the 20th week of gestation, making it a leading cause of maternal and fetal mortality worldwide. Despite extensive research, significant gaps remain in understanding the mechanisms underlying PE, contributing to the ineffectiveness of current prevention and treatment strategies. Consequently, premature cesarean sections often become the primary intervention to safeguard maternal and fetal health. Emerging evidence indicates that placental insufficiency, driven by molecular disturbances, plays a central role in the development of PE. Additionally, the maternal microbiome may be implicated in the pathomechanism of preeclampsia by secreting metabolites that influence maternal inflammation and oxidative stress, thereby affecting placental health. Given the limitations of pharmaceuticals during pregnancy due to potential risks to fetal development and concerns about teratogenic effects, nutraceuticals may provide safer alternatives. Nutraceuticals are food products or dietary supplements that offer health benefits beyond basic nutrition, including plant extracts or probiotics. Their historical use in traditional medicine has provided valuable insights into their safety and efficacy, including for pregnant women. This review will examine how the adoption of nutraceuticals can enhance dysregulated placental pathways, potentially offering benefits in the prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
| | | | | | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
33
|
Ma B, Li H, Huang Y, Guo Y, Xu C, Li W. Design, synthesis and activity screening of cedrol derivatives as small molecule JAK3 inhibitors. Bioorg Chem 2024; 152:107762. [PMID: 39222556 DOI: 10.1016/j.bioorg.2024.107762] [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: 05/28/2024] [Revised: 08/17/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The JAK-STAT signalling pathway is considered to be a significant role involved in the regulation of inflammatory diseases and immune responses, which indicate that specific inhibition of JAK-STAT pathway would be a potential key strategy for RA (Rheumatoid arthritis) treatment. Cedrol (CE), found from ginger by our group earlier, has been proven to play an excellent role in ameliorating RA via acting on JAK3. In this study, 27 new (1, 3-28), along with one known (2) derivatives of CE were synthesized by using chloroacetic acid and acryloyl chloride as intermediate ligands. In vitro, the inhibition effect on JAK kinases were performed using HTRF (Homogenous Time-Resolved Fluorescence) detection technology, which is more convenient and stable than traditional methods. The results compared with the secretion of LPS-induced p-JAK3 can better reflect the true kinase-selective effect of the compounds. Compound 22 was identified as a potent inhibitor to reduce the secretion of LPS-induced p-JAK3 with a dose-dependent manner. Given these results, compound 22 could serve as a favourable inhibitor of JAK3 for further research.
Collapse
Affiliation(s)
- Bingjing Ma
- Department of Traditional Chinese Material Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hua Li
- Department of Traditional Chinese Material Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuan Huang
- Liaoning Inspection, Examination & Certification Centre, Shenyang 110170, China
| | - Yaming Guo
- Department of Functional Food and Wine, Shenyang pharmaceutical university, Shenyang 110016, China
| | - Caizhu Xu
- Department of Functional Food and Wine, Shenyang pharmaceutical university, Shenyang 110016, China
| | - Wei Li
- Department of Functional Food and Wine, Shenyang pharmaceutical university, Shenyang 110016, China.
| |
Collapse
|
34
|
Sugumaran D, Yong ACH, Stanslas J. Advances in psoriasis research: From pathogenesis to therapeutics. Life Sci 2024; 355:122991. [PMID: 39153596 DOI: 10.1016/j.lfs.2024.122991] [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: 06/04/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Psoriasis is a chronic inflammatory condition affecting approximately 2 % to 3 % of the global population. The pathogenesis of psoriasis is complex, involving immune dysregulation, hyperproliferation and angiogenesis. It is a multifactorial disease which is influenced by genetic and environmental factors. The development of various therapeutic agents, such as JAK inhibitors, small molecules, and biologics with potential anti-psoriatic properties was possible with the vast understanding of the pathogenesis of psoriasis. Various signalling pathways, including NF-κB, JAK-STAT, S1P, PDE-4, and A3AR that are involved in the pathogenesis of psoriasis as well as the preclinical models utilised in the research of psoriasis have been highlighted in this review. The review also focuses on technological advancements that have contributed to a better understanding of psoriasis. Then, the molecules targeting the respective signalling pathways that are still under clinical trials or recently approved as well as the latest breakthroughs in therapeutic and drug delivery approaches that can contribute to the improvement in the management of psoriasis are highlighted in this review. This review provides an extensive understanding of the current state of research in psoriasis, giving rise to opportunities for researchers to discover future therapeutic breakthroughs and personalised interventions. Efficient treatment options for individuals with psoriasis can be achieved by an extensive understanding of pathogenesis, therapeutic agents, and novel drug delivery strategies.
Collapse
Affiliation(s)
- Dineshwar Sugumaran
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Audrey Chee Hui Yong
- Faculty of Pharmacy, Mahsa University, Bandar Saujana Putra, Jenjarom, Selangor, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutic Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
35
|
Aoki KC, Burnette C, Bartos S. Herpes Zoster and Venous Thromboembolism Following Upadacitinib Treatment for Severe Atopic Dermatitis. Cureus 2024; 16:e71007. [PMID: 39507180 PMCID: PMC11539937 DOI: 10.7759/cureus.71007] [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: 07/19/2024] [Accepted: 10/05/2024] [Indexed: 11/08/2024] Open
Abstract
New medications targeting the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway have been developed through emerging biologics research. However, due to documented adverse effects, including herpes zoster (HZ) and venous thromboembolism (VTE), an extensive patient workup and counseling are necessary before prescribing. We present the case of an 81-year-old patient with severe atopic dermatitis on upadacitinib, a selective JAK1 inhibitor, who developed HZ and VTE, requiring hospitalization. This study emphasizes the need for further research, continuous monitoring, and risk management for HZ and VTE in patients undergoing upadacitinib treatment, especially in high-risk populations.
Collapse
Affiliation(s)
- Kawaiola Cael Aoki
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, USA
| | - Colin Burnette
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, USA
| | | |
Collapse
|
36
|
Solimani F, Ghoreschi K. [Janus kinase inhibitors for skin disorders]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:781-790. [PMID: 39212722 DOI: 10.1007/s00105-024-05406-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Immune factors such as interferon‑ɣ and interleukin 4 belong to the group of cytokines that are dependent on type I/II receptors for their signal transmission. Upon activation, these receptors transmit their signal to the cell nucleus and, thus, modulate gene transcription via a signaling cascade consisting of Janus kinases (JAK). This family of four kinases (JAK 1, JAK 2, JAK 3, and tyrosine kinase 2 (TYK2)) subsequently activate members of the signal transducer and activator of transcription (STAT). This finding turned the JAK/STAT signaling pathway into a pharmacological target for the treatment of inflammatory diseases in which cytokines using type I/II receptors play a pathogenic role. In 2018, the European Medicines Agency (EMA) approved tofacitinib for the treatment of psoriatic arthritis. This was the first approval of a JAK/STAT pathway inhibitor for patients treated by dermatologists and rheumatologists. Since then, several new JAK inhibitors have been approved for dermatologic diseases such as atopic dermatitis, alopecia areata, vitiligo, and plaque-type psoriasis. In addition, JAK inhibitors are being investigated for the treatment of many other skin diseases. Thus, systemic JAK inhibitors complete the spectrum of immunotherapeutics with a broader immunological approach compared to monoclonal antibodies. The low molecular weight of JAK inhibitors enables the preparation of these drugs for both systemic and topical administration. Their utilization could represent a valuable alternative to topical steroids. The safety profile of JAK inhibitors must be taken into account. Possible long-term effects may become apparent in the next few years. This article describes both approved JAK inhibitors and relevant new JAK inhibitors that are promising candidates for approval as therapeutics in dermatology.
Collapse
Affiliation(s)
- Farzan Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Luisenstr. 2, 10117, Berlin, Deutschland.
- BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Deutschland.
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Luisenstr. 2, 10117, Berlin, Deutschland
| |
Collapse
|
37
|
Caballero-Mateos AM, Cañadas-de la Fuente GA. Game changer: How Janus kinase inhibitors are reshaping the landscape of ulcerative colitis management. World J Gastroenterol 2024; 30:3942-3953. [PMID: 39351053 PMCID: PMC11438661 DOI: 10.3748/wjg.v30.i35.3942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024] Open
Abstract
Recent advancements in the treatment landscape of ulcerative colitis (UC) have ushered in a new era of possibilities, particularly with the introduction of Janus kinase (JAK)-signal transducer and activator of transcription inhibitors. These novel agents offer a paradigm shift in UC management by targeting key signaling pathways involved in inflammatory processes. With approved JAK inhibitors (JAKis), such as tofacitinib, filgotinib, and upadacitinib, clinicians now have powerful tools to modulate immune responses and gene expression, potentially revolutionizing the treatment algorithm for UC. Clinical trials have demonstrated the efficacy of JAKis in inducing and maintaining remission, presenting viable options for patients who have failed conventional therapies. Real-world data support the use of JAKis not only as first-line treatments but also in subsequent lines of therapy, particularly in patients with aggressive disease phenotypes or refractory to biologic agents. The rapid onset of action and potency of JAKis have broadened the possibilities in the management strategies of UC, offering timely relief for patients with active disease and facilitating personalized treatment approaches. Despite safety concerns, including cardiovascular risks and infections, ongoing research and post-marketing surveillance will continue to refine our understanding of the risk-benefit profile of JAKis in UC management.
Collapse
|
38
|
Hsu CY, Mustafa MA, Moath Omar T, Taher SG, Ubaid M, Gilmanova NS, Nasrat Abdulraheem M, Saadh MJ, Athab AH, Mirzaei R, Karampoor S. Gut instinct: harnessing the power of probiotics to tame pathogenic signaling pathways in ulcerative colitis. Front Med (Lausanne) 2024; 11:1396789. [PMID: 39323474 PMCID: PMC11422783 DOI: 10.3389/fmed.2024.1396789] [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: 03/06/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) marked by persistent inflammation of the mucosal lining of the large intestine, leading to debilitating symptoms and reduced quality of life. Emerging evidence suggests that an imbalance of the gut microbiota plays a crucial role in UC pathogenesis, and various signaling pathways are implicated in the dysregulated immune response. Probiotics are live microorganisms that confer health benefits to the host, have attracted significant attention for their potential to restore gut microbial balance and ameliorate inflammation in UC. Recent studies have elucidated the mechanisms by which probiotics modulate these signaling pathways, often by producing anti-inflammatory molecules and promoting regulatory immune cell function. For example, probiotics can inhibit the nuclear factor-κB (NF-κB) pathway by stabilizing Inhibitor of kappa B alpha (IκBα), dampening the production of proinflammatory cytokines. Similarly, probiotics can modulate the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway, suppressing the activation of STAT1 and STAT3 and thus reducing the inflammatory response. A better understanding of the underlying mechanisms of probiotics in modulating pathogenic signaling pathways in UC will pave the way for developing more effective probiotic-based therapies. In this review, we explore the mechanistic role of probiotics in the attenuation of pathogenic signaling pathways, including NF-κB, JAK/STAT, mitogen-activated protein kinases (MAPKs), Wnt/β-catenin, the nucleotide-binding domain (NOD)-, leucine-rich repeat (LRR)- and pyrin domain-containing protein 3 (NLRP3) inflammasome, Toll-like receptors (TLRs), interleukin-23 (IL-23)/IL-17 signaling pathway in UC.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, United States
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, Imam Jaafar AL-Sadiq University, Baghdad, Iraq
- Department of Pathological Analyzes, College of Applied Sciences, University of Samarra, Samarra, Iraq
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq
| | - Sada Gh Taher
- Department of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammed Ubaid
- Department of MTL, Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Nataliya S. Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | - Aya H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
39
|
Vázquez-Jiménez LK, Rivera G, Juárez-Saldivar A, Ortega-Balleza JL, Ortiz-Pérez E, Jaime-Sánchez E, Paz-González A, Lara-Ramírez EE. Biological Evaluations and Computer-Aided Approaches of Janus Kinases 2 and 3 Inhibitors for Cancer Treatment: A Review. Pharmaceutics 2024; 16:1165. [PMID: 39339202 PMCID: PMC11435443 DOI: 10.3390/pharmaceutics16091165] [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: 07/24/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer remains one of the leading diseases of mortality worldwide. Janus kinases 2/3 (JAK2/3) have been considered a drug target for the development of drugs to treat different types of cancer. JAK2/3 play a critical role in innate immunity, inflammation, and hematopoiesis by mediating the signaling of numerous cytokines, growth factors, and interferons. The current focus is to develop new selective inhibitors for each JAK type. In this review, the current strategies of computer-aided studies, and biological evaluations against JAK2/3 are addressed. We found that the new synthesized JAK2/3 inhibitors are prone to containing heterocyclic aromatic rings such as pyrimidine, pyridine, and pyrazolo [3,4-d]pyrimidine. Moreover, inhibitors of natural origin derived from plant extracts and insects have shown suitable inhibitory capacities. Computer-assisted studies have shown the important features of inhibitors for JAK2/3 binding. Biological evaluations showed that the inhibition of the JAK receptor affects its related signaling pathway. Although the reviewed compounds showed good inhibitory capacity in vitro and in vivo, more in-depth studies are needed to advance toward full approval of cancer treatments in humans.
Collapse
Affiliation(s)
- Lenci K. Vázquez-Jiménez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City 03940, Mexico
| | - Gildardo Rivera
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
| | - Alfredo Juárez-Saldivar
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
| | - Jessica L. Ortega-Balleza
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City 03940, Mexico
| | - Eyra Ortiz-Pérez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
| | - Elena Jaime-Sánchez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City 03940, Mexico
| | - Alma Paz-González
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
| | - Edgar E. Lara-Ramírez
- Laboratorio de Biotecnología Farmacéutica, Centro de Biotecnología Genómica, Instituto Politécnico Nacional, Reynosa 88710, Mexico; (G.R.); (A.J.-S.); (J.L.O.-B.); (E.O.-P.); (E.J.-S.); (A.P.-G.)
| |
Collapse
|
40
|
Zhang X, Wang W, Dong G, Song Y, Zhai X, Sheng C. Discovery of a potent and selective JAK1-targeting PROTAC degrader with anti-tumor activities. Bioorg Med Chem Lett 2024; 109:129838. [PMID: 38838918 DOI: 10.1016/j.bmcl.2024.129838] [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: 03/12/2024] [Revised: 05/26/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Aberrant activation of the JAK-STAT pathway is evident in various human diseases including cancers. Proteolysis targeting chimeras (PROTACs) provide an attractive strategy for developing novel JAK-targeting drugs. Herein, a series of CRBN-directed JAK-targeting PROTACs were designed and synthesized utilizing a JAK1/JAK2 dual inhibitor-momelotinib as the warhead. The most promising compound 10c exhibited both good enzymatic potency and cellular antiproliferative effects. Western blot analysis revealed that compound 10c effectively and selectively degraded JAK1 in a proteasome-dependent manner (DC50 = 214 nM). Moreover, PROTAC 10c significantly suppressed JAK1 and its key downstream signaling. Together, compound 10c may serve as a novel lead compound for antitumor drug discovery.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Wei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, PR China
| | - Guoqiang Dong
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, PR China
| | - Yingqi Song
- School of Pharmaceutical Sciences, Hainan University, Haikou 570228, PR China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), 325 Guohe Road, Shanghai 200433, PR China.
| |
Collapse
|
41
|
O'Hare M, Guidon AC. Peripheral nervous system immune-related adverse events due to checkpoint inhibition. Nat Rev Neurol 2024; 20:509-525. [PMID: 39122934 DOI: 10.1038/s41582-024-01001-6] [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] [Accepted: 07/11/2024] [Indexed: 08/12/2024]
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy and are increasingly used to treat a wide range of oncological conditions, with dramatic benefits for many patients. Unfortunately, the resulting increase in T cell effector function often results in immune-related adverse events (irAEs), which can involve any organ system, including the central nervous system (CNS) and peripheral nervous system (PNS). Neurological irAEs involve the PNS in two-thirds of affected patients. Muscle involvement (immune-related myopathy) is the most common PNS irAE and can be associated with neuromuscular junction involvement. Immune-related peripheral neuropathy most commonly takes the form of polyradiculoneuropathy or cranial neuropathies. Immune-related myopathy (with or without neuromuscular junction involvement) often occurs along with immune-related myocarditis, and this overlap syndrome is associated with substantially increased mortality. This Review focuses on PNS adverse events associated with immune checkpoint inhibition. Underlying pathophysiological mechanisms are discussed, including antigen homology between self and tumour, epitope spreading and activation of pre-existing autoreactive T cells. An overview of current approaches to clinical management is provided, including cytokine-directed therapies that aim to decouple anticancer immunity from autoimmunity and emerging treatments for patients with severe (life-threatening) presentations.
Collapse
Affiliation(s)
- Meabh O'Hare
- Brigham and Women's Hospital, Division of Neuromuscular Medicine, Department of Neurology, Boston, MA, USA.
- Massachusetts General Hospital, Division of Neuromuscular Medicine, Department of Neurology, Boston, MA, USA.
| | - Amanda C Guidon
- Massachusetts General Hospital, Division of Neuromuscular Medicine, Department of Neurology, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Rzewińska A, Szlęk J, Dąbrowski D, Juszczyk E, Mróz K, Räikkönen H, Siven M, Wieczorek M, Dorożyński P. Development of a Formulation and In Vitro Evaluation of a Pulmonary Drug Delivery System for a Novel Janus Kinase (JAK) Inhibitor, CPL409116. Pharmaceutics 2024; 16:1157. [PMID: 39339194 PMCID: PMC11435004 DOI: 10.3390/pharmaceutics16091157] [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: 06/14/2024] [Revised: 08/13/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
The pursuit of targeted therapies for cytokine-dependent diseases has led to the discovery of Janus kinase (JAK) inhibitors, a promising class of drugs. Among them, CPL409116, a selective dual JAK and rho-associated protein kinase inhibitor (ROCK), has demonstrated potential for treating conditions such as pulmonary fibrosis exacerbated by the COVID-19 pandemic. This study investigated the feasibility of delivering CPL409116 via inhalation, with the aim of minimizing the systemic adverse effects associated with oral administration. Two micronization methods, jet milling and spray drying, were assessed for CPL409116, with spray drying chosen for its ability to produce an amorphous form of the compound. Moreover, parameters such as the mixing energy, drug load, and force control agent significantly influenced the fine particle fraction (FPF), a critical parameter for pulmonary drug delivery. This study provides insights into optimizing the formulation parameters to enhance the delivery efficiency of CPL409116 to the lungs, offering potential for improved therapeutic outcomes in cytokine-dependent pulmonary diseases.
Collapse
Affiliation(s)
- Aleksandra Rzewińska
- Finished Dosage Forms Department, Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
- Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warszawa, Poland
| | - Jakub Szlęk
- Chair and Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Damian Dąbrowski
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warszawa, Poland
| | - Ewelina Juszczyk
- Finished Dosage Forms Department, Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
| | - Katarzyna Mróz
- Finished Dosage Forms Department, Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
| | - Heikki Räikkönen
- Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, 00014 Helsinki, Finland
| | - Mia Siven
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, 00014 Helsinki, Finland
- Helsinki Institute of Sustainability Science HELSUS, University of Helsinki, 00014 Helsinki, Finland
| | - Maciej Wieczorek
- Research and Development Center, Celon Pharma S.A., Marymoncka 15, 05-052 Kazuń Nowy, Poland
| | - Przemysław Dorożyński
- Department of Drug Technology and Pharmaceutical Biotechnology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warszawa, Poland
- Department of Inorganic and Analytical Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
43
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [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: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
44
|
Zhang Y, Lin X, Xia L, Xiong S, Xia B, Xie J, Lin Y, Lin L, Wu P. Progress on the Anti-Inflammatory Activity and Structure-Efficacy Relationship of Polysaccharides from Medical and Edible Homologous Traditional Chinese Medicines. Molecules 2024; 29:3852. [PMID: 39202931 PMCID: PMC11356930 DOI: 10.3390/molecules29163852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Medicinal food varieties developed according to the theory of medical and edible homologues are effective at preventing and treating chronic diseases and in health care. As of 2022, 110 types of traditional Chinese medicines from the same source of medicine and food have been published by the National Health Commission. Inflammation is the immune system's first response to injury, infection, and stress. Chronic inflammation is closely related to many diseases such as atherosclerosis and cancer. Therefore, timely intervention for inflammation is the mainstay treatment for other complex diseases. However, some traditional anti-inflammatory drugs on the market are commonly associated with a number of adverse effects, which seriously affect the health and safety of patients. Therefore, the in-depth development of new safe, harmless, and effective anti-inflammatory drugs has become a hot topic of research and an urgent clinical need. Polysaccharides, one of the main active ingredients of medical and edible homologous traditional Chinese medicines (MEHTCMs), have been confirmed by a large number of studies to exert anti-inflammatory effects through multiple targets and are considered potential natural anti-inflammatory drugs. In addition, the structure of medical and edible homologous traditional Chinese medicines' polysaccharides (MEHTCMPs) may be the key factor determining their anti-inflammatory activity, which makes the underlying the anti-inflammatory effects of polysaccharides and their structure-efficacy relationship hot topics of domestic and international research. However, due to the limitations of the current analytical techniques and tools, the structures have not been fully elucidated and the structure-efficacy relationship is relatively ambiguous, which are some of the difficulties in the process of developing and utilizing MEHTCMPs as novel anti-inflammatory drugs in the future. For this reason, this paper summarizes the potential anti-inflammatory mechanisms of MEHTCMPs, such as the regulation of the Toll-like receptor-related signaling pathway, MAPK signaling pathway, JAK-STAT signaling pathway, NLRP3 signaling pathway, PI3K-AKT signaling pathway, PPAR-γ signaling pathway, Nrf2-HO-1 signaling pathway, and the regulation of intestinal flora, and it systematically analyzes and evaluates the relationships between the anti-inflammatory activity of MEHTCMPs and their structures.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiulian Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Li Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Suhui Xiong
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Bohou Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jingchen Xie
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yan Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Limei Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ping Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
45
|
Bissonnette R, Warren RB, Pinter A, Agner T, Gooderham M, Schuttelaar MLA, Crépy MN, Stingeni L, Serra-Baldrich E, Baranowski K, Korn S, Kurvits M, Plohberger U, Strange Vest N, Schliemann S. Efficacy and safety of delgocitinib cream in adults with moderate to severe chronic hand eczema (DELTA 1 and DELTA 2): results from multicentre, randomised, controlled, double-blind, phase 3 trials. Lancet 2024; 404:461-473. [PMID: 39033766 DOI: 10.1016/s0140-6736(24)01027-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Chronic hand eczema is a fluctuating, inflammatory, pruritic, often painful disease of hands and wrists that strongly impacts quality of life and occupational capabilities of patients. The aim of phase 3 DELTA 1 and DELTA 2 was to assess the efficacy and safety of twice-daily applications of the topical pan-Janus kinase inhibitor delgocitinib cream 20 mg/g versus cream vehicle in adults with moderate to severe chronic hand eczema. METHODS Both trials were randomised, double-blinded, and vehicle-controlled, with DELTA 1 being conducted at 53 trial centres in Canada, France, Germany, Italy, Poland, and the UK and DELTA 2 at 50 trial centres in Belgium, Canada, Denmark, Germany, the Netherlands, Poland, and Spain. Adults (aged ≥18 years) with moderate to severe chronic hand eczema were randomly assigned 2:1 to twice-daily delgocitinib cream 20 mg/g or cream vehicle for 16 weeks. The primary endpoint was Investigator's Global Assessment for Chronic Hand Eczema (IGA-CHE) treatment success at week 16, defined as IGA-CHE score of 0 (clear) or 1 (almost clear, defined as only barely perceptible erythema). Efficacy and safety were assessed in all patients who were exposed to trial treatment. These trials are registered with ClinicalTrials.gov, NCT04871711 and NCT04872101. FINDINGS Between May 10, 2021, and Oct 31, 2022, 487 patients (181 male and 306 female) were enrolled in DELTA 1; between May 25, 2021, and Jan 6, 2023, 473 patients (161 male and 312 female) were enrolled in DELTA 2. 325 patients in DELTA 1 and 314 in DELTA 2 were assigned to delgocitinib cream; 162 patients in DELTA 1 and 159 in DELTA 2 were assigned to cream vehicle. At week 16, a greater proportion of delgocitinib-treated patients versus cream vehicle patients had IGA-CHE treatment success (64 [20%] of 325 vs 16 [10%] of 162 in DELTA 1 and 91 [29%] of 313 vs 11 [7%] of 159 in DELTA 2; both trials p≤0·0055). The proportion of patients who reported adverse events was similar with delgocitinib (147 [45%] of 325 in DELTA 1 and 143 [46%] of 313 in DELTA 2) and the cream vehicle (82 [51%] of 162 in DELTA 1 and 71 [45%] of 159 in DELTA 2). Most frequent adverse events occurring in at least 2% of patients were similar in both treatment groups and included COVID-19 and nasopharyngitis. INTERPRETATION Overall, delgocitinib cream showed superior efficacy versus cream vehicle and was well tolerated over 16 weeks. These results support the clinical benefit of delgocitinib cream as a potential treatment option for patients with moderate to severe chronic hand eczema, who are unable to adequately control their disease with basic skin care practices and topical corticosteroids. FUNDING LEO Pharma.
Collapse
Affiliation(s)
| | - Richard B Warren
- Dermatology Centre, Northern Care Alliance NHS Foundation Trust, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Andreas Pinter
- Department of Dermatology, Venereology, and Allergology, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Tove Agner
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Melinda Gooderham
- Department of Medicine, Queens University, Kingston, ON, Canada; SKiN Centre for Dermatology and Probity Medical Research, Peterborough, ON, Canada
| | - Marie L A Schuttelaar
- University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Marie-Noëlle Crépy
- Department of Dermatology, University Hospital of Centre of Paris, Cochin Hospital, AP-HP, Paris, France; Department of Occupational and Environmental Diseases, University Hospital of Centre of Paris, Hôtel-Dieu Hospital, AP-HP, Paris, France
| | - Luca Stingeni
- Dermatology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Eshaq AM, Flanagan TW, Hassan SY, Al Asheikh SA, Al-Amoudi WA, Santourlidis S, Hassan SL, Alamodi MO, Bendhack ML, Alamodi MO, Haikel Y, Megahed M, Hassan M. Non-Receptor Tyrosine Kinases: Their Structure and Mechanistic Role in Tumor Progression and Resistance. Cancers (Basel) 2024; 16:2754. [PMID: 39123481 PMCID: PMC11311543 DOI: 10.3390/cancers16152754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Protein tyrosine kinases (PTKs) function as key molecules in the signaling pathways in addition to their impact as a therapeutic target for the treatment of many human diseases, including cancer. PTKs are characterized by their ability to phosphorylate serine, threonine, or tyrosine residues and can thereby rapidly and reversibly alter the function of their protein substrates in the form of significant changes in protein confirmation and affinity for their interaction with protein partners to drive cellular functions under normal and pathological conditions. PTKs are classified into two groups: one of which represents tyrosine kinases, while the other one includes the members of the serine/threonine kinases. The group of tyrosine kinases is subdivided into subgroups: one of them includes the member of receptor tyrosine kinases (RTKs), while the other subgroup includes the member of non-receptor tyrosine kinases (NRTKs). Both these kinase groups function as an "on" or "off" switch in many cellular functions. NRTKs are enzymes which are overexpressed and activated in many cancer types and regulate variable cellular functions in response to extracellular signaling-dependent mechanisms. NRTK-mediated different cellular functions are regulated by kinase-dependent and kinase-independent mechanisms either in the cytoplasm or in the nucleus. Thus, targeting NRTKs is of great interest to improve the treatment strategy of different tumor types. This review deals with the structure and mechanistic role of NRTKs in tumor progression and resistance and their importance as therapeutic targets in tumor therapy.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, George Washington University, Washington, DC 20052, USA;
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Sofie-Yasmin Hassan
- Department of Pharmacy, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Simeon Santourlidis
- Institute of Cell Therapeutics and Diagnostics, University Medical Center of Duesseldorf, 40225 Duesseldorf, Germany;
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Maryam O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Marcelo L. Bendhack
- Department of Urology, Red Cross University Hospital, Positivo University, Rua Mauá 1111, Curitiba 80030-200, Brazil;
| | - Mohammed O. Alamodi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (S.A.A.A.); (W.A.A.-A.); (M.O.A.); (M.O.A.)
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
47
|
Fan K, Gao Q, Cai C, Xie Y, Qi Z, Sun Z, Xie J, Gao J. Cloning and expression analysis of Janus activated kinase family genes from spotted seabass (Lateolabrax maculatus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 157:105169. [PMID: 38522714 DOI: 10.1016/j.dci.2024.105169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/22/2024] [Accepted: 03/22/2024] [Indexed: 03/26/2024]
Abstract
Janus kinases (JAKs) are important components of the JAK-STAT signaling pathway and play vital roles in innate immunity, autoimmune diseases, and inflammation. However, information about JAKs remains largely unknown in the spotted seabass, a fish species of Perciformes with great commercial value in the aquaculture industry. The aims of this study are to obtain the complete cDNA sequences of JAKs (JAK1, JAK2A, JAK2B, JAK3 and TYK2) from spotted seabass and to investigate their roles upon stimulation with lipopolysaccharides (LPS) and Edwardsiella tarda, using RT-PCR, PCR and qRT-PCR methods. All five JAK genes from the spotted seabass, each encode more than 1100 amino acids residues. JAK1 and JAK3 consist of 24 exons and 23 introns, whereas JAK2A, JAK2B and TYK2 consist of 23 exons and 22 introns. Furthermore, these five spotted seabass JAKs share high sequence identities with those of other fish species in protein domain analysis, synteny analysis, and phylogenetic analysis. Moreover, these five JAK genes were ubiquitously expressed in all tissues examined from healthy fish, and inducible expressions of JAKs were observed in the intestine, gill, head kidney, and spleen following LPS treatment or E. tarda infection. These findings indicate that all these JAK genes are involved in the antibacterial immunity of the spotted seabass and provide a basis for further understanding the mechanism of JAKs antibacterial response in the spotted sea bass.
Collapse
Affiliation(s)
- Ke Fan
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, 201306, China
| | - Qian Gao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, 201306, China.
| | - Chuanguo Cai
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, 201306, China
| | - Yushuai Xie
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, 201306, China
| | - Zhitao Qi
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu Province, 224051, China.
| | - Zhaosheng Sun
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, 201306, China
| | - Jiasong Xie
- School of Marine Sciences, Ningbo University, Zhejiang, 315211, China
| | - Jiaqi Gao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, 201306, China
| |
Collapse
|
48
|
Shan M, Zhao X, Sun P, Qu X, Cheng G, Qin LP. Revisiting Structure-activity Relationships: Unleashing the potential of selective Janus kinase 1 inhibitors. Bioorg Chem 2024; 149:107506. [PMID: 38833989 DOI: 10.1016/j.bioorg.2024.107506] [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: 03/20/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Janus kinases (JAKs), a kind of non-receptor tyrosine kinases, the function has been implicated in the regulation of cell proliferation, differentiation and apoptosis, immune, inflammatory response and malignancies. Among them, JAK1 represents an essential target for modulating cytokines involved in inflammation and immune function. Rheumatoid arthritis, atopic dermatitis, ulcerative colitis and psoriatic arthritis are areas where approved JAK1 drugs have been applied for the treatment. In the review, we provided a brief introduction to JAK1 inhibitors in market and clinical trials. The structures of high active JAK1 compounds (IC50 ≤ 0.1 nM) were highlighted, with primary focus on structure-activity relationship and selectivity. Moreover, the druggability processes of approved drugs and high active compounds were analyzed. In addition, the issues involved in JAK1 compounds clinical application as well as strategies to surmount these challenges, were discussed.
Collapse
Affiliation(s)
- Mengyi Shan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Xuan Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Peng Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Xinhao Qu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Gang Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China.
| | - Lu-Ping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China.
| |
Collapse
|
49
|
Kaur M, Misra S. Deucravacitinib: moderate-to-severe plaque psoriasis preventable? J Basic Clin Physiol Pharmacol 2024; 35:225-230. [PMID: 38976593 DOI: 10.1515/jbcpp-2023-0174] [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: 07/17/2023] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
Psoriasis is a persistent, inflammatory, and autoimmune condition that is difficult to treat. Estimates of the prevalence of psoriasis in people range from 0.27 % (95 % confidence interval 0.17 to 0.36) to 11.4 %, depending on factors such as age, sex, geography, ethnicity, genetics, and environmental factors. While systemic treatments are typically required for patients with moderate-to-severe instances of psoriasis, topical therapies are frequently effective for treating minor forms. In fact, phototherapy is frequently constrained by logistical considerations, and conventional systemic therapies are frequently avoided due to contraindications or the danger of adverse outcomes. In order to better serve the patient and achieve a greater level of quality of life, especially in order to sustain long-term efficacy, there is still a need for innovative therapies, which are always welcomed. Deucravacitinib is a first-in-class oral tyrosine kinase 2 (TYK2) inhibitor that is extremely selective. Through an allosteric mechanism, it stabilises an inhibitory connection between the regulatory and catalytic domains of TYK2's pseudokinase regulatory domain, which is catalytically inactive. This can be used to treat a variety of immune-mediated conditions, such as inflammatory bowel disease, lupus, psoriatic arthritis, and psoriasis. US-FDA has approved this drug on 9 September 2022 for the treatment of adults with moderate-to-severe plaque psoriasis who are candidates for systemic therapy or phototherapy. This article aims to review the current knowledge on the efficacy and safety of deucravacitinib for the management of psoriasis.
Collapse
Affiliation(s)
- Manmeet Kaur
- Department of Pharmacology, Kalpana Chawla Government Medical College, Karnal, Haryana, India
| | - Saurav Misra
- Department of Pharmacology, Kalpana Chawla Government Medical College, Karnal, Haryana, India
| |
Collapse
|
50
|
Andole S, Thumma G, Alavala RR, Gangarapu K. Field-based 3D-QSAR for tyrosine protein kinase JAK-2 inhibitors. J Biomol Struct Dyn 2024; 42:5321-5333. [PMID: 37409931 DOI: 10.1080/07391102.2023.2226723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/10/2023] [Indexed: 07/07/2023]
Abstract
The present work aimed to develop a Field-based 3D-QSAR model with existing JAK-2 inhibitors. The JAK-STAT pathway is known to play a role in the development of autoimmune diseases, including rheumatoid arthritis, ulcerative colitis, and Crohn's disease. Dysregulation of JAK-STAT is also linked to the development of myelofibrosis and other myeloproliferative diseases. JAK antagonists can be used in many areas of medicine. There are many compounds that already show inhibition of Jak-2. We have developed a Field-based 3D QSAR model which showed good correlation values (r2 0.884 and q2 0.67) with an external test set regression pred_r2 0.562. Various properties, such as electronegativity, electro positivity, hydrophobicity, and shape features, were studied under the activity atlas to determine the inhibitory potential of ligands. These were also identified as important structural features responsible for biological activity. We performed virtual screening based on the pharmacophore features of the co-crystal ligand (PDB ID: 3KRR) and a dataset of NPS was selected with a RMSD value less than 0.8. The developed 3D QSAR model was used to screen ligands and calculate the predicted JAK-2 inhibition activity (pKi). The results of the virtual screening were validated using molecular docking and molecular dynamics simulations. SNP1 (SN00154718) and SNP2 (SN00213825) showed binding affinity of -11.16 and -11.08 kcal/mol, respectively, which were very close to the crystal ligand of 3KRR, -11.67 kcal/mol. The RMSD plot of the protein-ligand complex of SNP1 and 3KRR showed stable interactions with an average RMSD of 2.89 Å. Thus, a statistically robust 3D QSAR model could reveal more inhibitors and aid in the design of novel JAK-2 inhibitors.
Collapse
Affiliation(s)
- Sowmya Andole
- School of Pharamcy, Anurag University, Venkatapur, Ghatkesar, Medchal-Malkajgiri district, Hyderabad, Telangana, India
| | - Gouthami Thumma
- University College of Pharmaceutical Sciences, Kakatiya University, Hanamkonda, Warangal, Telangana, India
| | - Rajasekhar Reddy Alavala
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, India
| | - Kiran Gangarapu
- School of Pharamcy, Anurag University, Venkatapur, Ghatkesar, Medchal-Malkajgiri district, Hyderabad, Telangana, India
| |
Collapse
|