1
|
Yang C, Merlin D. Unveiling Colitis: A Journey through the Dextran Sodium Sulfate-induced Model. Inflamm Bowel Dis 2024; 30:844-853. [PMID: 38280217 PMCID: PMC11063560 DOI: 10.1093/ibd/izad312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 01/29/2024]
Abstract
Animal models of inflammatory bowel disease (IBD) are valuable tools for investigating the factors involved in IBD pathogenesis and evaluating new therapeutic options. The dextran sodium sulfate (DSS)-induced model of colitis is arguably the most widely used animal model for studying the pathogenesis of and potential treatments for ulcerative colitis (UC), which is a primary form of IBD. This model offers several advantages as a research tool: it is highly reproducible, relatively easy to generate and maintain, and mimics many critical features of human IBD. Recently, it has also been used to study the role of gut microbiota in the development and progression of IBD and to investigate the effects of other factors, such as diet and genetics, on colitis severity. However, although DSS-induced colitis is the most popular and flexible model for preclinical IBD research, it is not an exact replica of human colitis, and some results obtained from this model cannot be directly applied to humans. This review aims to comprehensively discuss different factors that may be involved in the pathogenesis of DSS-induced colitis and the issues that should be considered when using this model for translational purposes.
Collapse
Affiliation(s)
- Chunhua Yang
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA, 30303, USA
- Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Digestive Disease Research Group, Georgia State University, Atlanta, GA, 30303, USA
- Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| |
Collapse
|
2
|
Temal-Laib T, Peixoto C, Desroy N, De Lemos E, Bonnaterre F, Bienvenu N, Picolet O, Sartori E, Bucher D, López-Ramos M, Roca Magadán C, Laenen W, Flower T, Mollat P, Bugaud O, Touitou R, Pereira Fernandes A, Lavazais S, Monjardet A, Borgonovi M, Gosmini R, Brys R, Amantini D, De Vos S, Andrews M. Optimization of Selectivity and Pharmacokinetic Properties of Salt-Inducible Kinase Inhibitors that Led to the Discovery of Pan-SIK Inhibitor GLPG3312. J Med Chem 2024; 67:380-401. [PMID: 38147525 PMCID: PMC10788895 DOI: 10.1021/acs.jmedchem.3c01428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/28/2023]
Abstract
Salt-inducible kinases (SIKs) SIK1, SIK2, and SIK3 are serine/threonine kinases and form a subfamily of the protein kinase AMP-activated protein kinase (AMPK) family. Inhibition of SIKs in stimulated innate immune cells and mouse models has been associated with a dual mechanism of action consisting of a reduction of pro-inflammatory cytokines and an increase of immunoregulatory cytokine production, suggesting a therapeutic potential for inflammatory diseases. Following a high-throughput screening campaign, subsequent hit to lead optimization through synthesis, structure-activity relationship, kinome selectivity, and pharmacokinetic investigations led to the discovery of clinical candidate GLPG3312 (compound 28), a potent and selective pan-SIK inhibitor (IC50: 2.0 nM for SIK1, 0.7 nM for SIK2, and 0.6 nM for SIK3). Characterization of the first human SIK3 crystal structure provided an understanding of the binding mode and kinome selectivity of the chemical series. GLPG3312 demonstrated both anti-inflammatory and immunoregulatory activities in vitro in human primary myeloid cells and in vivo in mouse models.
Collapse
Affiliation(s)
- Taouès Temal-Laib
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Nicolas Desroy
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Elsa De Lemos
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Natacha Bienvenu
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Olivier Picolet
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Eric Sartori
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Denis Bucher
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | | | - Wendy Laenen
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Thomas Flower
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Patrick Mollat
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Olivier Bugaud
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Robert Touitou
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | | | - Alain Monjardet
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Monica Borgonovi
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Romain Gosmini
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - David Amantini
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Steve De Vos
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Martin Andrews
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| |
Collapse
|
3
|
Babbe H, Sundberg TB, Tichenor M, Seierstad M, Bacani G, Berstler J, Chai W, Chang L, Chung DM, Coe K, Collins B, Finley M, Guletsky A, Lemke CT, Mak PA, Mathur A, Mercado-Marin EV, Metkar S, Raymond DD, Rives ML, Rizzolio M, Shaffer PL, Smith R, Smith J, Steele R, Steffens H, Suarez J, Tian G, Majewski N, Volak LP, Wei J, Desai PT, Ong LL, Koudriakova T, Goldberg SD, Hirst G, Kaushik VK, Ort T, Seth N, Graham DB, Plevy S, Venable JD, Xavier RJ, Towne JE. Identification of highly selective SIK1/2 inhibitors that modulate innate immune activation and suppress intestinal inflammation. Proc Natl Acad Sci U S A 2024; 121:e2307086120. [PMID: 38147543 PMCID: PMC10769863 DOI: 10.1073/pnas.2307086120] [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: 04/28/2023] [Accepted: 11/07/2023] [Indexed: 12/28/2023] Open
Abstract
The salt-inducible kinases (SIK) 1-3 are key regulators of pro- versus anti-inflammatory cytokine responses during innate immune activation. The lack of highly SIK-family or SIK isoform-selective inhibitors suitable for repeat, oral dosing has limited the study of the optimal SIK isoform selectivity profile for suppressing inflammation in vivo. To overcome this challenge, we devised a structure-based design strategy for developing potent SIK inhibitors that are highly selective against other kinases by engaging two differentiating features of the SIK catalytic site. This effort resulted in SIK1/2-selective probes that inhibit key intracellular proximal signaling events including reducing phosphorylation of the SIK substrate cAMP response element binding protein (CREB) regulated transcription coactivator 3 (CRTC3) as detected with an internally generated phospho-Ser329-CRTC3-specific antibody. These inhibitors also suppress production of pro-inflammatory cytokines while inducing anti-inflammatory interleukin-10 in activated human and murine myeloid cells and in mice following a lipopolysaccharide challenge. Oral dosing of these compounds ameliorates disease in a murine colitis model. These findings define an approach to generate highly selective SIK1/2 inhibitors and establish that targeting these isoforms may be a useful strategy to suppress pathological inflammation.
Collapse
Affiliation(s)
- Holger Babbe
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Thomas B. Sundberg
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Mark Tichenor
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Mark Seierstad
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Genesis Bacani
- Janssen Research and Development, LLC., San Diego, CA92121
| | - James Berstler
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Wenying Chai
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Leon Chang
- Janssen Research and Development, LLC., San Diego, CA92121
| | | | - Kevin Coe
- Janssen Research and Development, LLC., San Diego, CA92121
| | | | - Michael Finley
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Alexander Guletsky
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Christopher T. Lemke
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Puiying A. Mak
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Ashok Mathur
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Shailesh Metkar
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Donald D. Raymond
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | | | | | - Paul L. Shaffer
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Russell Smith
- Janssen Research and Development, LLC., San Diego, CA92121
| | | | - Ruth Steele
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Javier Suarez
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Gaochao Tian
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Nathan Majewski
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Jianmei Wei
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Prerak T. Desai
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Luvena L. Ong
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | | | - Gavin Hirst
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Virendar K. Kaushik
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Tatiana Ort
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Nilufer Seth
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Daniel B. Graham
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA02142
| | - Scott Plevy
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA02142
| | | |
Collapse
|
4
|
Zhu W, Liu X, Li Q, Gao F, Liu T, Chen X, Zhang M, Aliper A, Ren F, Ding X, Zhavoronkov A. Discovery of novel and selective SIK2 inhibitors by the application of AlphaFold structures and generative models. Bioorg Med Chem 2023; 91:117414. [PMID: 37467565 DOI: 10.1016/j.bmc.2023.117414] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023]
Abstract
Salt-inducible kinase 2 (SIK2) has been recognized as a potential target for anti-inflammation and anti-cancer therapy. In this paper, based on the binding pose of the reported compound (GLPG-3970, 3) with AlphaFold protein structure, a series of hinge cores were generated via AI-generative models (Chemistry42). After the molecular docking, synthesis, and biological evaluation, a hit molecule (7f) targeting SIK2 was obtained with a novel scaffold. Further SAR exploration led to the discovery of compound 8g with superior potency against SIK2 compared with the reported inhibitors. Furthermore, 8g also demonstrated excellent selectivity over other AMPK kinases, favorable in vitro ADMET profiles and decent cellular activities. This work provides an alternative approach to the discovery of novel and selective kinase inhibitors.
Collapse
Affiliation(s)
- Wei Zhu
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Xiaosong Liu
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Qi Li
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Feng Gao
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Tingting Liu
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Xiaojing Chen
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Alex Aliper
- Insilico Medicine AI Limited, Masdar City, Abu Dhabi 145748, UAE
| | - Feng Ren
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China
| | - Xiao Ding
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China.
| | - Alex Zhavoronkov
- Insilico Medicine Shanghai Ltd., Shanghai 201203, China; Insilico Medicine AI Limited, Masdar City, Abu Dhabi 145748, UAE.
| |
Collapse
|
5
|
Cai X, Wang L, Yi Y, Deng D, Shi M, Tang M, Li N, Wei H, Zhang R, Su K, Ye H, Chen L. Discovery of pyrimidine-5-carboxamide derivatives as novel salt-inducible kinases (SIKs) inhibitors for inflammatory bowel disease (IBD) treatment. Eur J Med Chem 2023; 256:115469. [PMID: 37178481 DOI: 10.1016/j.ejmech.2023.115469] [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: 12/05/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Salt-inducible kinases (SIKs) play a crucial role in inflammation process, acting as molecular switches that regulate the transformation of M1/M2 macrophages. HG-9-91-01 is a SIKs inhibitor with potent inhibitory activity against SIKs in the nanomolar range. However, its poor drug-like properties, including a rapid elimination rate, low in vivo exposure and high plasma protein binding rate, have hindered further research and clinical application. To improve the drug-like properties of HG-9-91-01, a series of pyrimidine-5-carboxamide derivatives were designed and synthesized through a molecular hybridization strategy. The most promising compound 8h was obtained with favorable activity and selectivity on SIK1/2, excellent metabolic stability in human liver microsome, enhanced in vivo exposure and suitable plasma protein binding rate. Mechanism research showed that compound 8h significantly up-regulated the expression of anti-inflammatory cytokine IL-10 and reduced the expression of pro-inflammatory cytokine IL-12 in bone marrow-derived macrophages. Furthermore, it significantly elevated expression of cAMP response element-binding protein (CREB) target genes IL-10, c-FOS and Nurr77. Compound 8h also induced the translocation of CREB-regulated transcriptional coactivator 3 (CRTC3) and elevated the expression of LIGHT, SPHK1 and Arginase 1. Additionally, compound 8h demonstrated excellent anti-inflammatory effects in a DSS-induced colitis model. Generally, this research indicated that compound 8h has the potential to be developed as an anti-inflammatory drug candidate.
Collapse
Affiliation(s)
- Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lun Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyao Yi
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Dexin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mingsong Shi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haoche Wei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Chengdu Zenitar Biomedical Technology Co., Ltd, Chengdu, China.
| |
Collapse
|
6
|
Cheng Y, Li J, Wang L, Wu X, Li Y, Xu M, Li Q, Huang J, Zhao T, Yang Z, Zhang H, Zuo L, Zhang X, Geng Z, Wang Y, Song X, Jun Z. Eriocalyxin B ameliorated Crohn's disease-like colitis by restricting M1 macrophage polarization through JAK2/STAT1 signalling. Eur J Pharmacol 2023:175876. [PMID: 37391008 DOI: 10.1016/j.ejphar.2023.175876] [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: 01/28/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND AND AIMS M1 polarization of macrophages in the intestine is an important maintenance factor of the inflammatory response in Crohn's disease (CD). Eriocalyxin B (EriB) is a natural medicine that antagonizes inflammation. Our study aimed to determine the effects of EriB on CD-like colitis in mice, as well as the possible mechanism. METHODS 2,4,6-trinitrobenzene sulfonic acid (TNBS) mice and Il-10-/- mice were used as CD animal models, and the therapeutic effect of EriB on CD-like colitis in mice was addressed by the disease activity index (DAI) score, weight change, histological analysis and flow cytometry assay. To assess the direct role of EriB in regulating macrophage polarization, bone marrow-derived macrophages (BMDMs) were induced to M1 or M2 polarization separately. Molecular docking simulations and blocking experiments were performed to explore the potential mechanisms by which EriB regulates the macrophage polarization. RESULTS EriB treatment reduced body weight loss, DAI score and histological score, demonstrating the improvement of colitis symptoms in mice. In vivo and in vitro experiments both showed that EriB decreased the M1 polarization of macrophages, and suppressed the release of proinflammatory cytokines (IL-1β, TNF-α and IL-6) in mouse colons and BMDMs. The activation of Janus kinase 2/signal transducer and activator of transcription 1 (JAK2/STAT1) signals could be inhibited by EriB, which may be related to the regulation of EriB on M1 polarization. CONCLUSIONS EriB inhibits the M1 polarization of macrophages by attenuating the JAK2/STAT1 pathway, which partially explains the potential mechanism by which EriB ameliorates colitis in mice, and provides a new regimen for the clinical treatment of CD.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Blood Transfusion, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lian Wang
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaopei Wu
- Bengbu Medical College, Bengbu, Anhui, China
| | - Yuetong Li
- Bengbu Medical College, Bengbu, Anhui, China
| | - Mengyu Xu
- Bengbu Medical College, Bengbu, Anhui, China
| | - Qingqing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ju Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Tianhao Zhao
- Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Zi Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Lugen Zuo
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijun Geng
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xue Song
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| | - Zhang Jun
- Department of Blood Transfusion, First Affiliated Hospital of Bengbu Medical College, Bengbu, China.
| |
Collapse
|
7
|
Huang J, Fan H, Chen YM, Wang CN, Guan W, Li WY, Shi TS, Chen WJ, Zhu BL, Liu JF, Jiang B. The salt-inducible kinases inhibitor HG-9-91-01 exhibits antidepressant-like actions in mice exposed to chronic unpredictable mild stress. Neuropharmacology 2023; 227:109437. [PMID: 36702294 DOI: 10.1016/j.neuropharm.2023.109437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/24/2023]
Abstract
Major depressive disorder is a frequently occurring neuropsychiatric disorder throughout the world. However, the limited and delayed therapeutic efficacy of monoaminergic medications has led to intensive research efforts to develop novel antidepressants. We have previously demonstrated that hippocampal salt-inducible kinase 2 (SIK2) plays a role in the pathogenesis of depression via regulating the downstream CREB-regulated transcription coactivator 1 (CRTC1)-cAMP response element-binding protein (CREB)-brain derived neurotrophic factor (BDNF) pathway. HG-9-91-01 is a potent and selective inhibitor of salt-inducible kinases (SIKs). The present study aims to explore whether HG-9-91-01 has antidepressant-like actions in male C57BL/6J mice. The chronic unpredictable mild stress (CUMS) model of depression, various behavioral tests, western blotting, co-immunoprecipitation, immunofluorescence, stereotactic infusion, and viral-mediated genetic knockdown were used together. It was found that hippocampal infusion of HG-9-91-01 induced significant antidepressant-like effects in the CUMS model, accompanied with preventing the enhancement of CUMS on the hippocampal SIK2 expression and cytoplasmic translocation of CRTC1. HG-9-91-01 treatment also reversed the decreasing effects of CUMS on the BDNF signaling cascade and adult neurogenesis in the hippocampus. Moreover, the antidepressant-like actions of HG-9-91-01 in mice required the hippocampal CRTC1-CREB-BDNF pathway. In conclusion, HG-9-91-01 has potential of being a novel antidepressant candidate.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Cheng-Niu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei-Yu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Tian-Shun Shi
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei-Jia Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Bao-Lun Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jian-Feng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
8
|
Huang D, Chen P, Huang G, Sun H, Luo X, He C, Chen F, Wang Y, Zeng C, Su L, Zeng X, Lu J, Li S, Huang D, Gao H, Cao M. Salt-inducible kinases inhibitor HG-9-91-01 targets RIPK3 kinase activity to alleviate necroptosis-mediated inflammatory injury. Cell Death Dis 2022; 13:188. [PMID: 35217652 PMCID: PMC8881470 DOI: 10.1038/s41419-022-04633-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/18/2022] [Accepted: 02/10/2022] [Indexed: 12/24/2022]
Abstract
Receptor-interacting protein kinase 3 (RIPK3) functions as a central regulator of necroptosis, mediating signaling transduction to activate pseudokinase mixed lineage kinase domain-like protein (MLKL) phosphorylation. Increasing evidences show that RIPK3 contributes to the pathologies of inflammatory diseases including multiple sclerosis, infection and colitis. Here, we identified a novel small molecular compound Salt-inducible Kinases (SIKs) inhibitor HG-9-91-01 inhibiting necroptosis by targeting RIPK3 kinase activity. We found that SIKs inhibitor HG-9-91-01 could block TNF- or Toll-like receptors (TLRs)-mediated necroptosis independent of SIKs. We revealed that HG-9-91-01 dramatically decreased cellular activation of RIPK3 and MLKL. Meanwhile, HG-9-91-01 inhibited the association of RIPK3 with MLKL and oligomerization of downstream MLKL. Interestingly, we found that HG-9-91-01 also trigger RIPK3-RIPK1-caspase 1-caspase 8-dependent apoptosis, which activated cleavage of GSDME leading to its dependent pyroptosis. Mechanistic studies revealed that SIKs inhibitor HG-9-91-01 directly inhibited RIPK3 kinase activity to block necroptosis and interacted with RIPK3 and recruited RIPK1 to activate caspases leading to cleave GSDME. Importantly, mice pretreated with HG-9-91-01 showed resistance to TNF-induced systemic inflammatory response syndrome. Consistently, HG-9-91-01 treatment protected mice against Staphylococcus aureus-mediated lung damage through targeting RIPK3 kinase activity. Overall, our results revealed that SIKs inhibitor HG-9-91-01 is a novel inhibitor of RIPK3 kinase and a potential therapeutic target for the treatment of necroptosis-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Dongxuan Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Pengfei Chen
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Guoqing Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Huimin Sun
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Xiaohua Luo
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Chaowen He
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Fei Chen
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Yong Wang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Changchun Zeng
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Lianhui Su
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China
| | - Xiaobin Zeng
- The State Key Lab of Respiratory Disease, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, 510182, China
| | - Jiachun Lu
- The State Key Lab of Respiratory Disease, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, 510182, China
| | - Shiyue Li
- The State Key Lab of Respiratory Disease, The First Affiliated Hospital, The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, 510182, China
| | - Dongsheng Huang
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China.
| | - Hanchao Gao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China.
| | - Mengtao Cao
- Department of Respiratory Medicine, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, 518110, China.
| |
Collapse
|