1
|
Wen T, Liu T, Chen H, Liu Q, Shen X, Hu Q. Demethylzeylasteral alleviates inflammation and colitis via dual suppression of NF-κB and STAT3/5 by targeting IKKα/β and JAK2. Int Immunopharmacol 2024; 142:113260. [PMID: 39340986 DOI: 10.1016/j.intimp.2024.113260] [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: 07/15/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a common inflammatory bowel disease and a risk factor of colorectal cancer. Demethylzeylasteral (DZT), a bioactive component mainly isolated from Tripterygium wilfordii, has been shown to inhibit inflammation and cancer. However, its anti-UC function and molecular mechanisms have not been well characterized. This study aims to explore the therapeutic effect and functional targets of demethylzeylasteral against UC. METHODS RT-qPCR, Western blot and ELISA were used to detect the generation of pro-inflammatory cytokines and chemokines in murine macrophage cells. Luciferase reporter gene, Western blot, pull-down, CETSA, DARTS, and virtual docking were employed to detect the anti-inflammatory targets and molecular mechanisms of demethylzeylasteral. The anti-inflammatory and anti-colitis effects of demethylzeylasteral were further determined in DSS-challenged mice. RESULTS In vitro, demethylzeylasteral inhibited NO and PGE2 production by suppressing the mRNA and protein expression of iNOS and COX-2, and suppressed the mRNA expression of TNF-α, IL-1β, IL-6, MCP-1, CXCL9, and CXCL10 in RAW264.7 macrophages stimulated by LPS/IFNγ. Furthermore, demethylzeylasteral was not only capable of inhibiting IKKα/β-NF-κB activation, but also able to block JAKs-STAT3/5 activation in LPS/INFγ-incubated RAW264.7 cells or DSS-exposed colon tissues of mice. Mechanistically, demethylzeylasteral was found to directly bind to IKKα/β and JAK2 kinases, leading to inactivation of pro-inflammatory signaling cascades and reduced generation of cytokines and chemokines. In vivo, oral administration of demethylzeylasteral significantly attenuated DSS-induced colitis, which was mainly manifested as mitigated symptoms of colitis, colonic mucosal barrier damage, and colonic inflammation. CONCLUSION We demonstrated that demethylzeylasteral alleviated UC pathology by blocking NF-κB and STAT3/5 pathways via targeting IKKα/β and JAK2 kinases, raising the possibility that demethylzeylasteral could act as a candidate for the treatment of UC.
Collapse
Affiliation(s)
- Tian Wen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Ting Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Hongqing Chen
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qi Liu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Xiaofei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Qiongying Hu
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
2
|
Cao J, Yu X, Liu J, Fu J, Wang B, Wu C, Zhang S, Chen H, Wang Z, Xu Y, Sui T, Chang J, Cao X. Ruxolitinib improves the inflammatory microenvironment, restores glutamate homeostasis, and promotes functional recovery after spinal cord injury. Neural Regen Res 2024; 19:2499-2512. [PMID: 38526286 PMCID: PMC11090442 DOI: 10.4103/nrr.nrr-d-23-01863] [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: 11/12/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00030/figure1/v/2024-03-08T184507Z/r/image-tiff The inflammatory microenvironment and neurotoxicity can hinder neuronal regeneration and functional recovery after spinal cord injury. Ruxolitinib, a JAK-STAT inhibitor, exhibits effectiveness in autoimmune diseases, arthritis, and managing inflammatory cytokine storms. Although studies have shown the neuroprotective potential of ruxolitinib in neurological trauma, the exact mechanism by which it enhances functional recovery after spinal cord injury, particularly its effect on astrocytes, remains unclear. To address this gap, we established a mouse model of T10 spinal cord contusion and found that ruxolitinib effectively improved hindlimb motor function and reduced the area of spinal cord injury. Transcriptome sequencing analysis showed that ruxolitinib alleviated inflammation and immune response after spinal cord injury, restored EAAT2 expression, reduced glutamate levels, and alleviated excitatory toxicity. Furthermore, ruxolitinib inhibited the phosphorylation of JAK2 and STAT3 in the injured spinal cord and decreased the phosphorylation level of nuclear factor kappa-B and the expression of inflammatory factors interleukin-1β, interleukin-6, and tumor necrosis factor-α. Additionally, in glutamate-induced excitotoxicity astrocytes, ruxolitinib restored EAAT2 expression and increased glutamate uptake by inhibiting the activation of STAT3, thereby reducing glutamate-induced neurotoxicity, calcium influx, oxidative stress, and cell apoptosis, and increasing the complexity of dendritic branching. Collectively, these results indicate that ruxolitinib restores glutamate homeostasis by rescuing the expression of EAAT2 in astrocytes, reduces neurotoxicity, and effectively alleviates inflammatory and immune responses after spinal cord injury, thereby promoting functional recovery after spinal cord injury.
Collapse
Affiliation(s)
- Jiang Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiao Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jingcheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jiaju Fu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Binyu Wang
- Department of Trauma Surgery, Subei People’s Hospital of Jiangsu, Clinical Medical College of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Chaoqin Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sheng Zhang
- Department of Orthopedics, Zhongda Hospital, Southeast University, Nanjing, Jiangsu Province, China
| | - Hongtao Chen
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Zi Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yinyang Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Sui
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jie Chang
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiaojian Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
3
|
Han ZQ, Wen LN. Tofacitinib for ulcerative colitis: A promising treatment option. World J Gastroenterol 2024; 30:4386-4392. [PMID: 39494100 PMCID: PMC11525864 DOI: 10.3748/wjg.v30.i40.4386] [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: 07/28/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 10/16/2024] Open
Abstract
A single center retrospective clinical study revealed the efficacy and safety of tofacitinib in the treatment of ulcerative colitis (UC). This study has clinical reference value but also has some limitations. Previous studies, including this clinical trial, have shown that tofacitinib could be a promising treatment option for UC, but further clinical research is required to prove this point.
Collapse
Affiliation(s)
- Zong-Qiang Han
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital, Beijing 102211, China
| | - Li-Na Wen
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| |
Collapse
|
4
|
Pei CS, Hou XO, Ma ZY, Tu HY, Qian HC, Li Y, Li K, Liu CF, Ouyang L, Liu JY, Hu LF. α-Synuclein disrupts microglial autophagy through STAT1-dependent suppression of Ulk1 transcription. J Neuroinflammation 2024; 21:275. [PMID: 39462396 PMCID: PMC11515151 DOI: 10.1186/s12974-024-03268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Autophagy dysfunction in glial cells is implicated in the pathogenesis of Parkinson's disease (PD). The previous study reported that α-synuclein (α-Syn) disrupted autophagy in cultured microglia. However, the mechanism of microglial autophagy dysregulation is poorly understood. METHODS Two α-Syn-based PD models were generated via AAV-mediated α-Syn delivery into the mouse substantia nigra and striatal α-Syn preformed fibril (PFF) injection. The levels of microglial UNC-51-like kinase 1 (Ulk1) and other autophagy-related genes in vitro and in PD mice, as well as in the peripheral blood mononuclear cells of PD patients and healthy controls, were determined via quantitative PCR, western blotting and immunostaining. The regulatory effect of signal transducer and activator of transcription 1 (STAT1) on Ulk1 transcription was determined via a luciferase reporter assay and other biochemical studies and was verified through Stat1 knockdown or overexpression. The effect of α-Syn on glial STAT1 activation was assessed by immunohistochemistry and western blotting. Changes in microglial status, proinflammatory molecule expression and dopaminergic neuron loss in the nigrostriatum of PD and control mice following microglial Stat1 conditional knockout (cKO) or treatment with the ULK1 activator BL-918 were evaluated by immunostaining and western blotting. Motor behaviors were determined via open field tests, rotarod tests and balance beam crossing. RESULTS The transcription of microglial ULK1, a kinase that controls autophagy initiation, decreased in both in vitro and in vivo PD mouse models. STAT1 plays a critical role in suppressing Ulk1 transcription. Specifically, Stat1 overexpression downregulated Ulk1 transcription, while Stat1 knockdown increased ULK1 expression, along with an increase in LC3II and a decrease in the SQSTM1/p62 protein. α-Syn PFF caused toll-like receptor 4-dependent activation of STAT1 in microglia. Ablation of Stat1 alleviated the decrease in microglial ULK1 expression and disruption of autophagy caused by α-Syn PFF. Importantly, the ULK1 activator BL-918 and microglial Stat1 cKO attenuated neuroinflammation, dopaminergic neuronal damage and motor defects in PD models. CONCLUSIONS These findings reveal a novel mechanism by which α-Syn impairs microglial autophagy and indicate that targeting STAT1 or ULK1 may be a therapeutic strategy for PD.
Collapse
Affiliation(s)
- Chong-Shuang Pei
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Xiao-Ou Hou
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zhen-Yuan Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Hai-Yue Tu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Hai-Chun Qian
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Li
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jun-Yi Liu
- Department of Neurology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China.
| | - Li-Fang Hu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China.
- Institute of Neuroscience, Soochow University, 199 Ren-Ai Road, Suzhou Industrial Park, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
5
|
Zeng L, Wang Y, Shen J, Wei X, Wu Y, Chi X, Zheng X, Yu X, Shi Y, Liu W. TIPE2 aggravates experimental colitis and disrupts intestinal epithelial barrier integrity by activating JAK2/STAT3/SOCS3 signal pathway. Exp Cell Res 2024; 443:114287. [PMID: 39426612 DOI: 10.1016/j.yexcr.2024.114287] [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: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Ulcerative colitis (UC) is a chronic relapsing and progressive inflammatory disease of the colon. TIPE2 is a negative regulator of innate and adaptive immunity that maintains immune homeostasis. We found that TIPE2 was highly expressed in mucosa of mice with colitis. However, the role of TIPE2 in colitis remains unclear. We induced colitis in mice with dextran sulfate sodium (DSS) and treated them with TIPE2, and investigated the inflammatory activity of the colon in vivo by cytokines detection and histopathological analyses. We also measured inflammatory alteration and tight junctions induced by DSS in vitro. The results demonstrated that administration of TIPE2 promoted the severity of colitis in mice and human colon epithelial cells. Furthermore, TIPE2 aggravated intestinal epithelial barrier dysfunction by decreasing the expression of the tight junction proteins Occludin, Claudin-1 and ZO-1. In addition, TIPE2 exacerbated intestinal inflammatory response by inhibiting the expression of SOCS3, remarkably activating JAK2/STAT3 signaling pathway, and increasing the translocation of phosphorylated STAT3 into the nucleus. Silencing of TIPE2 attenuated the DSS-induced activation of JAK2/STAT3, thereby rescuing epithelial inflammatory injury and restoring barrier dysfunction. These results indicate that TIPE2 augments experimental colitis and disrupted the integrity of the intestinal epithelial barrier by activating the JAK2/STAT3/SOCS3 signaling pathway.
Collapse
Affiliation(s)
- Lingli Zeng
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuping Wang
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jiaxin Shen
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xujin Wei
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yilong Wu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xintong Chi
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xueyan Zheng
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xing Yu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Shi
- Department of Gastroenterology, The First Afiiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| | - Wenming Liu
- Endoscopy Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China; Endoscopy Center, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
6
|
Zhao X, Zhang Y, Wang P, Liu K, Zheng Y, Wen J, Wang K, Wen X. Layer by layer self-assembled hyaluronic acid nanoarmor for the treatment of ulcerative colitis. J Nanobiotechnology 2024; 22:633. [PMID: 39420343 PMCID: PMC11488142 DOI: 10.1186/s12951-024-02933-3] [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: 05/27/2024] [Accepted: 10/13/2024] [Indexed: 10/19/2024] Open
Abstract
Natural compound-based treatments provide innovative ways for ulcerative colitis therapy. However, poor targeting and rapid degradation curtail its application, which needs to be addressed. Inspired by biomacromolecule-based materials, we have developed an orally administrated nanoparticle (GBP@HA NPs) using bovine serum albumin as a carrier for polyphenol delivery. The system synergizes galactosylated bovine serum albumin with two polyphenols, epigallocatechin gallate and tannic acid, which is then encased in "nanoarmor" of ε-Polylysine and hyaluronic acid to boost its stability and targeting. Remarkably, the nanoarmor demonstrated profound therapeutic effects in both acute and chronic mouse models of ulcerative colitis, mitigating disease symptoms via multiple mechanisms, regulating inflammation related factors and exerting a modulatory impact on gut microbiota. Further mechanistic investigations indicate that GBP@HA NPs may act through several pathways, including modulation of Keap1-Nrf2 and NF-κB signaling, as well as Caspase-1-dependent pyroptosis. Consequently, this novel armored nanotherapy promotes the way for enhanced polyphenol utilization in ulcerative colitis treatment research.
Collapse
Affiliation(s)
- Xinxin Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuchen Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Pengchong Wang
- Department of Pharmacy, Shaanxi Provincial People's Hospital, Xi'an Shaanxi, 710068, China
| | - Kailai Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yunhe Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jinpeng Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ke Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Xiaopeng Wen
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
7
|
Saleem M, Aden LA, Mutchler AL, Basu C, Ertuglu LA, Sheng Q, Penner N, Hemnes AR, Park JH, Ishimwe JA, Laffer CL, Elijovich F, Wanjalla CN, de la Visitacion N, Kastner PD, Albritton CF, Ahmad T, Haynes AP, Yu J, Graber MK, Yasmin S, Wagner KU, Sayeski PP, Hatzopoulos AK, Gamazon ER, Bick AG, Kleyman TR, Kirabo A. Myeloid-Specific JAK2 Contributes to Inflammation and Salt Sensitivity of Blood Pressure. Circ Res 2024; 135:890-909. [PMID: 39263750 PMCID: PMC11466692 DOI: 10.1161/circresaha.124.323595] [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: 02/16/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Salt sensitivity of blood pressure (SSBP), characterized by acute changes in blood pressure with changes in dietary sodium intake, is an independent risk factor for cardiovascular disease and mortality in people with and without hypertension. We previously found that elevated sodium concentration activates antigen-presenting cells (APCs), resulting in high blood pressure, but the mechanisms are unknown. Here, we hypothesized that APC-specific JAK2 (Janus kinase 2) through STAT3 (signal transducer and activator of transcription 3) and SMAD3 (small mothers against decapentaplegic homolog 3) contributes to SSBP. METHODS We performed bulk or single-cell transcriptomic analyses following in vitro monocytes exposed to high salt and in vivo high sodium treatment in humans using a rigorous salt-loading/depletion protocol to phenotype SSBP. We also used a myeloid cell-specific CD11c+ JAK2 knockout mouse model and measured blood pressure with radiotelemetry after N-omega-nitro-L-arginine-methyl ester and a high salt diet treatment. We used flow cytometry for immunophenotyping and measuring cytokine levels. Fluorescence in situ hybridization and immunohistochemistry were performed to spatially visualize the kidney's immune cells and cytokine levels. Echocardiography was performed to assess cardiac function. RESULTS We found that high salt treatment upregulates gene expression of the JAK/STAT/SMAD pathway while downregulating inhibitors of this pathway, such as suppression of cytokine signaling and cytokine-inducible SH2, in human monocytes. Expression of the JAK2 pathway genes mirrored changes in blood pressure after salt loading and depletion in salt-sensitive but not salt-resistant humans. Ablation of JAK2, specifically in CD11c+ APCs, attenuated salt-induced hypertension in mice with SSBP. Mechanistically, we found that SMAD3 acted downstream of JAK2 and STAT3, leading to increased production of highly reactive isolevuglandins and proinflammatory cytokine IL (interleukin)-6 in renal APCs, which activate T cells and increase production of IL-17A, IL-6, and TNF-α (tumor necrosis factor-alpha). CONCLUSIONS Our findings reveal the APC JAK2 signaling pathway as a potential target for the diagnosis and treatment of SSBP in humans.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Luul A Aden
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Ashley L Mutchler
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Chitra Basu
- Department of Medicine, Division of Genetic Medicine (C.B., E.R.G.), Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Division of Cardiovascular Medicine (C.B., A.K.H.), Vanderbilt University Medical Center, Nashville, TN
| | - Lale A Ertuglu
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Quanhu Sheng
- Department of Biostatistics (Q.S.), Vanderbilt University Medical Center, Nashville, TN
| | - Niki Penner
- Division of Allergy, Pulmonary, and Critical Care Medicine (N.P., A.R.H.)
| | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine (N.P., A.R.H.)
| | - Jennifer H Park
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Cheryl L Laffer
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | | | - Celestine N Wanjalla
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Nestor de la Visitacion
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Paul D Kastner
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Claude F Albritton
- School of Graduate Studies, Meharry Medical College, Nashville, TN (C.F.A.)
| | - Taseer Ahmad
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Punjab, Pakistan (T.A.)
| | - Alexandria P Haynes
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Justin Yu
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Meghan K Graber
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Sharia Yasmin
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Kay-Uwe Wagner
- Wayne State University, Department of Oncology and Tumor Biology Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI (K.-U.W.)
| | - Peter P Sayeski
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville (P.P.S.)
| | - Antonis K Hatzopoulos
- Department of Medicine, Division of Cardiovascular Medicine (C.B., A.K.H.), Vanderbilt University Medical Center, Nashville, TN
| | - Eric R Gamazon
- Department of Medicine, Division of Genetic Medicine (C.B., E.R.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Alexander G Bick
- Division of Genetic Medicine (A.G.B.), Vanderbilt University Medical Center, Nashville, TN
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, PA (T.R.K.)
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology (M.S., L.A.A., A.L.M., L.A.E., J.H.P., J.A.I., C.L.L., C.N.W., N.d.l.V., P.D.K., T.A., A.P.H., J.Y., M.K.G., S.Y., A.K.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Center for Immunobiology (A.K.)
- Vanderbilt Institute for Infection, Immunology and Inflammation (A.K.)
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN (A.K.)
| |
Collapse
|
8
|
Liang Y, Li Y, Lee C, Yu Z, Chen C, Liang C. Ulcerative colitis: molecular insights and intervention therapy. MOLECULAR BIOMEDICINE 2024; 5:42. [PMID: 39384730 PMCID: PMC11464740 DOI: 10.1186/s43556-024-00207-w] [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: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by abdominal pain, diarrhea, rectal bleeding, and weight loss. The pathogenesis and treatment of UC remain key areas of research interest. Various factors, including genetic predisposition, immune dysregulation, and alterations in the gut microbiota, are believed to contribute to the pathogenesis of UC. Current treatments for UC include 5-aminosalicylic acids, corticosteroids, immunosuppressants, and biologics. However, study reported that the one-year clinical remission rate is only around 40%. It is necessary to prompt the exploration of new treatment modalities. Biologic therapies, such as anti-TNF-α monoclonal antibody and JAK inhibitor, primarily consist of small molecules targeting specific pathways, effectively inducing and maintaining remission. Given the significant role of the gut microbiota, research into intestinal microecologics, such as probiotics and prebiotics, and fecal microbiota transplantation (FMT) shows promising potential in UC treatment. Additionally, medicinal herbs, such as chili pepper and turmeric, used in complementary therapy have shown promising results in UC management. This article reviews recent findings on the mechanisms of UC, including genetic susceptibility, immune cell dynamics and cytokine regulation, and gut microbiota alterations. It also discusses current applications of biologic therapy, herbal therapy, microecologics, and FMT, along with their prospects and challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yang Li
- Department of Respiratory, Sichuan Integrative Medicine Hospital, Chengdu, 610042, China
| | - Chehao Lee
- Department of Traditional Chinese Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziwei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chongli Chen
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chao Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
9
|
Dasgupta S. Systems Biology and Machine Learning Identify Genetic Overlaps Between Lung Cancer and Gastroesophageal Reflux Disease. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:492-503. [PMID: 39269895 DOI: 10.1089/omi.2024.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
One Health and planetary health place emphasis on the common molecular mechanisms that connect several complex human diseases as well as human and planetary ecosystem health. For example, not only lung cancer (LC) and gastroesophageal reflux disease (GERD) pose a significant burden on planetary health, but also the coexistence of GERD in patients with LC is often associated with a poor prognosis. This study reports on the genetic overlaps between these two conditions using systems biology-driven bioinformatics and machine learning-based algorithms. A total of nine hub genes including IGHV1-3, COL3A1, ITGA11, COL1A1, MS4A1, SPP1, MMP9, MMP7, and LOC102723407 were found to be significantly altered in both LC and GERD as compared with controls and with pathway analyses suggesting a significant association with the matrix remodeling pathway. The expression of these genes was validated in two additional datasets. Random forest and K-nearest neighbor, two machine learning-based algorithms, achieved accuracies of 89% and 85% for distinguishing LC and GERD, respectively, from controls using these hub genes. Additionally, potential drug targets were identified, with molecular docking confirming the binding affinity of doxycycline to matrix metalloproteinase 7 (binding affinity: -6.8 kcal/mol). The present study is the first of its kind that combines in silico and machine learning algorithms to identify the gene signatures that relate to both LC and GERD and promising drug candidates that warrant further research in relation to therapeutic innovation in LC and GERD. Finally, this study also suggests upstream regulators, including microRNAs and transcription factors, that can inform future mechanistic research on LC and GERD.
Collapse
Affiliation(s)
- Sanjukta Dasgupta
- Department of Biotechnology, Center for Multidisciplinary Research and Innovations, Brainware University, Barasat, India
| |
Collapse
|
10
|
Chen Z, Jiang P, Su D, Zhao Y, Zhang M. Therapeutic inhibition of the JAK-STAT pathway in the treatment of inflammatory bowel disease. Cytokine Growth Factor Rev 2024; 79:1-15. [PMID: 39179485 DOI: 10.1016/j.cytogfr.2024.07.008] [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: 06/19/2024] [Revised: 07/19/2024] [Accepted: 07/20/2024] [Indexed: 08/26/2024]
Abstract
Inflammatory bowel disease (IBD) encompasses a group of non-specific chronic intestinal inflammatory conditions of unclear etiology. The current treatment and long-term management primarily involve biologics. Nevertheless, some patients experience treatment failure or intolerance to biologics [1], making these patients a primary focus of IBD research. The Janus kinase (JAK)-Signal Transducers and Activator of Transcription (STAT) signal transduction pathway is crucial to the regulation of immune and inflammatory responses [2], and plays an important role in the pathogenesis of IBD. JAK inhibitors alleviate IBD by suppressing the transmission of JAK-STAT signaling pathway. As the first small-molecule oral inhibitor for IBD, JAK inhibitors greatly improved the treatment of IBD and have demonstrated significant efficacy, with tofacitinib and upadacitinib being approved for the treatment of ulcerative colitis (UC) [3]. JAK inhibitors can effectively alleviate intestinal inflammation in IBD patients who have failed to receive biologics, which may bring new treatment opportunities for refractory IBD patients. This review aims to elucidate the crucial roles of JAK-STAT signal transduction pathway in IBD pathogenesis, examine its role in various cell types within IBD, and explore the research progress of JAK inhibitors as therapeutic agents, paving the road for new IBD treatment strategies.
Collapse
Affiliation(s)
- Zihan Chen
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Ping Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing 210093, China
| | - Dan Su
- FUJIFILM Diosynth Biotechnologies, Watertown, 02472, MA, United States
| | - Yu Zhao
- University of Chicago, Pritzker School of Molecular Engineering, Chicago, IL, 60637
| | - Mingming Zhang
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China; Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China.
| |
Collapse
|
11
|
Odah T, Karime C, Desai A, Picco MF, Kinnucan JA, Hashash JG, Farraye FA. Response to Upadacitinib in Patients with Inflammatory Bowel Disease Previously Treated with Tofacitinib. Dig Dis Sci 2024; 69:3911-3919. [PMID: 39251560 DOI: 10.1007/s10620-024-08630-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND AND AIMS Upadacitinib is an oral selective Janus kinase (JAK) inhibitor approved in the United States for ulcerative colitis (UC) and Crohn's disease (CD). However, data regarding its use following prior treatment with the JAK inhibitor tofacitinib is sparse. As such, we aimed to evaluate the effectiveness of upadacitinib therapy following tofacitinib exposure. METHODS This is a multicenter retrospective study of patients with confirmed diagnosis of UC or CD who received upadacitinib after prior treatment with tofacitinib. The primary outcome of interest was patient-reported clinical improvement at first follow-up. Secondary outcome included discontinuation of corticosteroids, change in Mayo Endoscopic Score (MES) and change in inflammatory marker levels. RESULTS A total of 31 patients met the inclusion criteria. Following upadacitinib initiation, 80.6% (25/31) of patients had clinical improvement, including 92.3% (24/26) of those with UC and 20% (1/5) of those with CD. Of the patients initially requiring systemic corticosteroid therapy, 80% (12/15) were able to discontinue corticosteroids. Individual mean change of fecal calprotectin was a decrease of 501.5 mcg/g ± 608.6 (P value = 0.01) while C-reactive protein decreased on average by 14.8 mg/L ± 25.3 (P value = 0.02) compared to when patients were on tofacitinib, with significant changes observed in the UC cohort. In patients with UC, individual MES after initiating upadacitinib decreased compared to prior to tofacitinib discontinuation (P value = 0.04). CONCLUSION Our study demonstrates that upadacitinib therapy in patients with prior tofacitinib exposure is associated with clinical improvement and a decrease in objective markers of inflammation in patients with UC.
Collapse
Affiliation(s)
- Tarek Odah
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, 4500 San Pablo Rd S., Jacksonville, FL, 32224, USA.
| | - Christian Karime
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Aakash Desai
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, 4500 San Pablo Rd S., Jacksonville, FL, 32224, USA
| | - Michael F Picco
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, 4500 San Pablo Rd S., Jacksonville, FL, 32224, USA
| | - Jami A Kinnucan
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, 4500 San Pablo Rd S., Jacksonville, FL, 32224, USA
| | - Jana G Hashash
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, 4500 San Pablo Rd S., Jacksonville, FL, 32224, USA
| | - Francis A Farraye
- Division of Gastroenterology and Hepatology, Inflammatory Bowel Disease Center, Mayo Clinic, 4500 San Pablo Rd S., Jacksonville, FL, 32224, USA
| |
Collapse
|
12
|
Chen X, Lou Y, Zhou F, Shi D, Liu X, Tao F. Identification of novel indolinone derivatives as CTSC inhibitors to treat inflammatory bowel disease by modulating inflammatory factors. Eur J Med Chem 2024; 280:116914. [PMID: 39383651 DOI: 10.1016/j.ejmech.2024.116914] [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: 08/05/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024]
Abstract
Novel inflammatory bowel disease (IBD) therapeutic drugs, mainly biologics that neutralize pro-inflammatory factors and janus kinase inhibitors that inhibit cytokine-mediated signal transduction, face problems including low efficacy rates, limited therapeutic benefits, and infection risks. It is an important task to find proteins that broadly regulate a variety of cytokines and to develop corresponding drugs. Cathepsin C (CTSC) mediates neutrophil-related inflammatory, participates in the recruitment and activation of inflammatory cells, and regulates cytokines levels, and is considered an ideal target for IBD treatment. In this study, starting from the in-house molecule, through medicinal chemistry and target-based design, a novel CTSC inhibitor B22 with IBD therapeutic efficacy was discovered. In vitro target verification and mechanism study indicated that B22 inhibit CTSC activity by binding to S2 pocket and S1 site, further inhibiting downstream serine protease activity. In addition, B22 exhibited anti-inflammatory activity and regulated various cytokines levels. In vivo studies highlighted B22 bears acceptable toxicity and suitable pharmacokinetic properties, and displays anti-inflammatory activity in IBD model. In conclusion, B22 is a potential anti-inflammatory molecule for IBD by targeting CTSC and deserves further research.
Collapse
Affiliation(s)
- Xing Chen
- School of Public Health, Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230032, PR China
| | - Yan Lou
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China
| | - Feilong Zhou
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China
| | - Daxing Shi
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China
| | - Xinhua Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, PR China.
| | - Fangbiao Tao
- School of Public Health, Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, 230032, PR China.
| |
Collapse
|
13
|
Su S, Liu T, Zheng JY, Wu HC, Keng VW, Zhang SJ, Li XX. Huang Lian Jie Du decoction attenuated colitis via suppressing the macrophage Csf1r/Src pathway and modulating gut microbiota. Front Immunol 2024; 15:1375781. [PMID: 39391314 PMCID: PMC11464287 DOI: 10.3389/fimmu.2024.1375781] [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: 01/24/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction Ulcerative colitis, a subtype of chronic inflammatory bowel disease (IBD), is characterized by relapsing colonic inflammation and ulcers. The traditional Chinese herbal formulation Huang Lian Jie Du (HLJD) decoction is used clinically to treat diarrhea and colitis. However, the mechanisms associated with the effects of treatment remain unclear. This study aims to elucidate the molecular mechanistic effects of HLJD formulation on colitis. Methods Chronic colitis in mice was induced by adding 1% dextran sulfate sodium (DSS) to their drinking water continuously for 8 weeks, and HLJD decoction at the doses of 2 and 4 g/kg was administered orally to mice daily from the second week until experimental endpoint. Stool consistency scores, blood stool scores, and body weights were recorded weekly. Disease activity index (DAI) was determined before necropsy, where colon tissues were collected for biochemical analyses. In addition, the fecal microbiome of treated mice was characterized using 16S rRNA amplicon sequencing. Results HLJD decoction at doses of 2 and 4 g/kg relieved DSS-induced chronic colitis in mice by suppressing inflammation through compromised macrophage activity in colonic tissues associated with the colony-stimulating factor 1 receptor (Csf1r)/Src pathway. Furthermore, the HLJD formula could modify the gut microbiota profile by decreasing the abundance of Bacteroides, Odoribacter, Clostridium_sensu_stricto_1, and Parasutterella. In addition, close correlations between DAI, colon length, spleen weight, and gut microbiota were identified. Discussion Our findings revealed that the HLJD formula attenuated DSS-induced chronic colitis by reducing inflammation via Csf1r/Src-mediated macrophage infiltration, as well as modulating the gut microbiota profile.
Collapse
Affiliation(s)
- Shan Su
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ting Liu
- Department of Pharmacy, Shenzhen Children’s Hospital, Shenzhen, China
| | - Jia-Yi Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Hai-Cui Wu
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Vincent W. Keng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Shi-Jie Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiao-Xiao Li
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
14
|
Nicolò S, Faggiani I, Errico C, D'Amico F, Parigi TL, Danese S, Ungaro F. Translational characterization of immune pathways in inflammatory bowel disease: insights for targeted treatments. Expert Rev Clin Immunol 2024:1-18. [PMID: 39313992 DOI: 10.1080/1744666x.2024.2400300] [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: 01/15/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The pathogenesis of inflammatory bowel disease (IBD) involves the dysregulation of multiple inflammatory pathways. The understanding of these mechanisms allows their selective targeting for therapeutic purposes. The discovery of Tumor Necrosis Factor-alpha's (TNF-α) role in mucosal inflammation ushered an exciting new era of drug development which now comprises agents targeting multiple pro-inflammatory signaling pathways, integrins, and leukocyte trafficking regulators. AREA COVERED This review provides an overview of the main molecular players of IBD, their translation into therapeutic targets and the successful development of the advanced agents modulating them. We combine basic science with clinical trials data to present a critical review of both the successful and failed drug development programs. A PubMed literature search was conducted to delve into the available literature and clinical trials. EXPERT OPINION The treatment landscape for IBD has rapidly expanded, particularly with the development of biologics targeting TNF-α, integrins, and S1P modulators, as well as newer agents such as IL-12/IL-23 inhibitors and JAK inhibitors, offering robust efficacy and safety profiles. However, challenges persist in understanding and effectively treating difficult-to-treat IBD, highlighting the need for continued research to uncover novel therapeutic targets and optimize patient outcomes.
Collapse
Affiliation(s)
- Sabrina Nicolò
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ilaria Faggiani
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Carmela Errico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ferdinando D'Amico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
15
|
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
|
16
|
Wang T, Wu W, Zhang X, Gan B, Zhou Y, Cheng X. Tofacitinib treatment for plaque psoriasis and psoriatic arthritis: A meta-analysis of randomised controlled trials. Indian J Dermatol Venereol Leprol 2024; 0:1-8. [PMID: 39361844 DOI: 10.25259/ijdvl_14_2024] [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: 01/04/2024] [Accepted: 06/26/2024] [Indexed: 10/05/2024]
Abstract
Objectives Tofacitinib is used as an oral Janus-associated kinase (JAK) inhibitor acting on JAK1 and JAK3, in treating psoriatic disease. However, there is still no consensus on the optimal dosage and duration of tofacitinib. In this study, we aimed to evaluate the effects of tofacitinib in treating psoriatic disease. Methods and Materials A literature search was done utilising Cochrane library, Medline, EMBASE, Wiley Online library, Web of Science and BIOSIS Previews through December 18, 2022. We performed a meta-analysis of published original studies to assess the impact of tofacitinib in plaque psoriasis or psoriatic arthritis therapy based on seven randomised controlled trials (RCTs) involving 2,672 patients (receiving tofacitinib) and 853 controls (receiving placebo). Results Compared with placebo, the treatment of 5 mg twice-daily (BID) tofacitinib for 12 weeks is sufficient to significantly alleviate the main clinical manifestations of psoriasis [≥75% decrease in Psoriasis Area and Severity Index score (PASI 75): Risk ratio (RR)=4.38 (95% Confidence interval (CI) 2.51 to 7.64); ≥90% decrease in PASI score (PASI 90): RR=21.68 (95% CI 4.20 to 111.85); Physician's Global Assessment of 'clear' or 'almost clear' (PGA 0/1): RR=3.93 (95%CI 3.03 to 5.09)]. Interestingly, there was no significant difference in improvement in PGA 0/1 with 5 mg BID tofacitinib given for 16 weeks when compared with 5 mg BID tofacitinib for 12 weeks [RR=1.11 (95%CI 0.98 to 1.25)]. Additionally, the 5 mg BID tofacitinib for 16 weeks treatment schedule significantly increased the incidence of upper respiratory tract infection (URTI) [RR=1.89 (95%CI 1.06 to 3.38)] as compared to 5 mg BID tofacitinib for 12 weeks treatment schedule [RR=1.15 (95%CI 0.60 to 2.20)]. Conclusion The 5 mg BID tofacitinib for 12 weeks treatment significantly improved psoriasis without causing too many specific adverse events. This indicated that tofacitinib is an effective treatment plan for psoriatic disease by reasonably controlling dosage and dosing time.
Collapse
Affiliation(s)
- Tao Wang
- Department of Sexual Medicine/Traditional Chinese Medicine, Fifth People's Hospital of Shunde District (Longjiang Hospital of Shunde District), Foshan, Guangdong, China
| | - Wei Wu
- Department of Medical Science, Shunde Polytechnic, Foshan, Guangdong, China
| | - Xiaoqing Zhang
- Department of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, China
| | - Bin Gan
- Department of Respiratory Medicine, Fifth People's Hospital of Shunde District (Longjiang Hospital of Shunde District), Foshan, Guangdong, China
| | - Yanfang Zhou
- Department of Basic Medical Sciences, Guangdong Medical University, Dongguan, Guangdong, China
| | - Xiaoyan Cheng
- Department of Traditional Chinese Medicine, The Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong, China
| |
Collapse
|
17
|
Xv Y, Feng Y, Lin J. CXCR1 and CXCR2 are potential neutrophil extracellular trap-related treatment targets in ulcerative colitis: insights from Mendelian randomization, colocalization and transcriptomic analysis. Front Immunol 2024; 15:1425363. [PMID: 39328405 PMCID: PMC11424450 DOI: 10.3389/fimmu.2024.1425363] [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: 04/29/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Objectives There is already substantial evidence indicating that neutrophil extracellular trap (NET) formation contributes to the inflammatory cascade in ulcerative colitis (UC). However, the precise regulatory mechanisms governing this process remain elusive. This study aimed to determine the role of NET-related genes in UC and reveal possible mechanisms. Methods Employing a two-sample MR methodology, we investigated the correlations between NET-associated genes (NRGs) and UC with summary data derived from a genome-wide association study (12,366 cases vs. 33,609 controls) and FinnGen (8,279 cases vs. 261,098 controls). The main analysis employed the inverse variance weighted method, supplemented by the MR-Egger method and weighted median method. Sensitivity analysis was conducted to rule out the interference of heterogeneity and pleiotropy among utilized instrument variables. The colocalization analysis was used to determine whether the identified NRGs and UC shared casual variants. Cross-tissue expression analysis was performed to characterize the expression patterns of target NRGs, while multi-gene correlation analysis and GSEA analysis were conducted to explore the mechanisms by which target NRGs promote UC and NET formation. Immunohistochemistry was used to validate the protein expression of target NRGs in the colon tissue of UC patients. Results After the validation of two datasets, seven NRGs were associated with the risk of UC. The higher expression of ITGB2 was associated with increased UC risk, while the expression of CXCR1, CXCR2, IRAK4, MAPK3, SIGLEC14, and SLC22A4 were inversely associated with UC risk. Colocalization analysis supported the correlation between CXCR1/2 and UC risk. Expression analysis indicated that CXCR1/2 were down-regulated in peripheral blood, but up-regulated in colon tissue. GSEA analysis and correlation analysis indicated that CXCR1/2 promoted UC and NET formation through neutrophil chemotaxis and PAD4-mediated pathways, separately. Immunohistochemical results confirmed the high expression of CXCR1/2 in colon tissues of UC patients. Conclusions Our study identified CXCR1/2 as candidate targets in UC among all NRGs through multi-method argumentation, providing new insights of the regulation mechanisms of NET formation in the pathogenesis of UC.
Collapse
Affiliation(s)
- Yichuan Xv
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyi Feng
- Department of Rheumatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang Lin
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
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
|
19
|
Xu X, Peng J, Wang N, Ocansey DKW, Zhang X, Mao F. hucMSC-Ex alleviates inflammatory bowel disease in mice by enhancing M2-type macrophage polarization via the METTL3-Slc37a2-YTHDF1 axis. Cell Biol Toxicol 2024; 40:74. [PMID: 39259386 PMCID: PMC11390928 DOI: 10.1007/s10565-024-09921-1] [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] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) have emerged as a new treatment strategy for inflammatory bowel disease (IBD) due to their immunoregulatory function. N6-methyladenosine (m6A) plays a crucial role in regulating intestinal immunity, especially in IBD where macrophages play an important role, although its mechanism is not yet fully understood. From this perspective, this research aimed to evaluate the effect of hucMSC-Ex on m6A modification of macrophages in IBD. In the process of alleviating inflammation, hucMSC-Ex promotes macrophage polarization toward the M2 type and regulates intracellular m6A levels by upregulating the expression of m6A "Writer" METTL3 and "Reader" YTHDF1. Solute Carrier Family 37 Member 2 (Slc37a2) was identified by Methylation RNA immunoprecipitation sequencing as the target molecule of the hucMSC-Ex. Mechanically, hucMSC-Ex promoted the binding of METTL3 to the Slc37a2 mRNA complex, and enhanced the binding of Slc37a2 to YTHDF1 to upregulate the intracellular expression of Slc37a2, thereby attenuating the pro-inflammatory function of macrophage. This study confirms the modulatory role of hucMSC-Ex on the m6A modification of macrophages in IBD, providing a new scientific basis for the treatment of IBD with hucMSC-Ex.
Collapse
Affiliation(s)
- Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China
- Clinical Laboratory, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221000, Jiangsu, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212300, Jiangsu, P.R. China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, CC0959347, Cape Coast, Ghana
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China.
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China.
| |
Collapse
|
20
|
Baik S, Kim H, Lee Y, Kang T, Shin K, Song C, Park OK, Kang B, Lee N, Kim D, Choi SH, Kim SH, Soh M, Hyeon T, Kim CK. Orally Deliverable Iron-Ceria Nanotablets for Treatment of Inflammatory Bowel Disease. Adv Healthc Mater 2024:e2401994. [PMID: 39235381 DOI: 10.1002/adhm.202401994] [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: 05/30/2024] [Revised: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Ceria-based nanoparticles are versatile in treating various inflammatory diseases, but their feasibility in clinical translation is undermined by safety concerns and a limited delivery system. Meanwhile, the idiopathic nature of inflammatory bowel disease (IBD) calls for a wider variety of therapeutics via moderation of the intestinal immune system. In this regard, the synthesis and oral formulation of iron-ceria nanoparticles (CF NPs) with enhanced nanozymic activity and lower toxicity risk than conventional ceria-based nanoparticles are reported. CF NPs are clustered in calcium phosphate (CaP) and coated with a pH-responsive polymer to provide the enteric formulation of iron-ceria nanotablets (CFNT). CFNT exhibits a marked alleviative efficacy in the dextran sodium sulfate (DSS)-induced enterocolitis model in vivo by modulating the pro-inflammatory behavior of innate immune cells including macrophages and neutrophils, promoting anti-inflammatory cytokine profiles, and downregulating key transcription factors of inflammatory pathways.
Collapse
Affiliation(s)
- Seungmin Baik
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Hyunmin Kim
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Department of Medical Science, BK21 Plus KUMS Graduate Program, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Yunjung Lee
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Taegyu Kang
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Kwangsoo Shin
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Changyeong Song
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Ok Kyu Park
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Byeonggeun Kang
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nohyun Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Advanced Materials Engineering, Kookmin University, Seoul, 02707, Republic of Korea
| | - Dokyoon Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15558, Republic of Korea
| | - Seung Hong Choi
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio Inc., Seoul, 08826, Republic of Korea
| | - Seung Han Kim
- Department of Gastroenterology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Min Soh
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Advanced Pharmaceutical Technology, HyeonTechNBio Inc., Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- School of Chemical and Biological Engineering and Institute of Chemical Process, Seoul National University, Seoul, 08826, Republic of Korea
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Chi Kyung Kim
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, 08308, Republic of Korea
- Department of Medical Science, BK21 Plus KUMS Graduate Program, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| |
Collapse
|
21
|
Honan AM, Jacobsen GE, Drum H, Vazquez EN, Quintero MA, Deshpande AR, Sussman DA, Kerman DH, Damas OM, Proksell S, Van der Jeught K, Abreu MT, Chen Z. Stromal-Like Cells Are Found in Peripheral Blood of Patients With Inflammatory Bowel Disease and Correlate With Immune Activation State. Clin Transl Gastroenterol 2024; 15:e1. [PMID: 38829958 PMCID: PMC11421714 DOI: 10.14309/ctg.0000000000000721] [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: 02/20/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
INTRODUCTION Recent studies have identified a critical role of stromal-immune cell interactions in immunity and immune tolerance. Transcriptomic profiling has implicated stromal cells in immune-mediated disorders including the 2 common forms of inflammatory bowel disease (IBD), Crohn's disease (CD), and ulcerative colitis (UC). Stromal-immune interactions may edify inflammatory state and the development of IBD-related complications such as fibrosis, yet the lack of protein markers has hampered studying stromal-immune perturbation. METHODS In this study, we designed a 40-color spectral flow cytometry assay to characterize hematopoietic and nonhematopoietic cells in intestinal biopsies and matched blood samples from patients with CD or UC. RESULTS We identified circulating stromal-like cells that are significantly more abundant in IBD blood samples than in healthy controls. Those cells expressed podoplanin (PDPN), a commonly used marker for fibroblasts, and they were associated with activated and memory T and B cells and altered natural killer cell, monocyte, and macrophage populations. PDPN + cells in the blood correlated with PDPN + cells in the colon. Principal component analysis distinctly separated healthy blood samples from IBD blood samples, with stromal-like cells and B-cell subtypes dominating the IBD signature; Pearson correlation detected an association between PDPN + stromal-like cells and B-cell populations in IBD blood and gut biopsies. DISCUSSION These observations suggest that PDPN + cells in the blood may serve as a biomarker of IBD. Understanding the relationship between stromal cells and immune cells in the intestine and the blood may provide a window into disease pathogenesis and insight into therapeutic targets for IBD.
Collapse
Affiliation(s)
- Amanda M. Honan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Gillian E. Jacobsen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Hannah Drum
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Emily N. Vazquez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Maria A. Quintero
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
| | - Amar R. Deshpande
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
| | - Daniel A. Sussman
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David H. Kerman
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
| | - Oriana M. Damas
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
| | - Siobhan Proksell
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
| | - Kevin Van der Jeught
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Maria T. Abreu
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
22
|
Lu X, Xu Y, Li X, Wang J, Wang L, Hu X, Fan H, Pang F, Li X, Pan X, He W, Li J, Dai Z. Selective STAT3 inhibitor STX-0119 alleviates osteoarthritis progression by modulating the STAT3/PPARγ signaling pathway. Biochem Pharmacol 2024; 227:116420. [PMID: 38996934 DOI: 10.1016/j.bcp.2024.116420] [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: 06/08/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Osteoarthritis (OA), characterized by chronic pain, significantly affects the quality of life of affected individuals. Key factors in OA pathogenesis include cartilage degradation and inflammation. Signal transducer and activator of transcription 3 (STAT3), a member of the STAT protein family, plays a pivotal role in mediating inflammation. STX-0119 has been verified as a small molecular compound that can specifically inhibit STAT3. However, the efficacy of STX-0119 in the treatment of OA remains to be evaluated. Therefore, the aim of this study was to explore the therapeutic effects and molecular mechanisms of STX-0119 in the treatment of OA. We found that the expression of phosphorylated STAT3 is upregulated in human OA cartilage as well as in the cartilage of a mouse model of OA. In vivo, joint injection of STX-0119 into OA mice alleviated cartilage degeneration without affecting the subchondral bone. Additionally, STX-0119 could inhibit the phosphorylation of STAT3 in the cartilage. In vitro, STX-0119 suppressed inflammatory responses in chondrocytes and promoted anabolic metabolism in an interleukin-1β-induced chondrocyte inflammation model. Additionally, the results of transcriptome sequencing and lentiviral infection assays demonstrated that in chondrocytes, STX-0119 induces the upregulation of peroxisome proliferators-activated receptor gamma (PPARγ) expression by inhibiting STAT3 phosphorylation. Finally, in ex vivo cultures of human cartilage samples, STX-0119 was reaffirmed to inhibit cartilage degeneration via the STAT3/PPARγ signaling pathway. Together, our findings support the potential of STX-0119 for development as a therapeutic agent targeting STAT3 for the treatment of OA.
Collapse
Affiliation(s)
- Xuanyuan Lu
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian, China; Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Yangjun Xu
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Xinhuo Li
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Jing Wang
- Department of Pediatric, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Lei Wang
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Xujun Hu
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Honghui Fan
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Fei Pang
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Xin Li
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Xiaoyu Pan
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian, China
| | - Wei He
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Jianlei Li
- Department of Orthopedic, Shaoxing People's Hospital, Shaoxing 312000, Zhejiang, China
| | - Zhangsheng Dai
- Department of Orthopedic, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, Fujian, China.
| |
Collapse
|
23
|
Liu H, Guan L, Su X, Zhao L, Shu Q, Zhang J. A broken network of susceptibility genes in the monocytes of Crohn's disease patients. Life Sci Alliance 2024; 7:e202302394. [PMID: 38925865 PMCID: PMC11208737 DOI: 10.26508/lsa.202302394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Genome-wide association studies have identified over 200 genetic loci associated with inflammatory bowel disease; however, the mechanism of such a large amount of susceptibility genes remains uncertain. In this study, we integrated bioinformatics analysis and two independent single-cell transcriptome datasets to investigate the expression network of 232 susceptibility genes in Crohn's disease (CD) patients and healthy controls. The study revealed that most of the susceptibility genes are specifically and strictly expressed in the monocytes of the human intestinal tract. The susceptibility genes established a network within the monocytes of health control. The robustness of a gene network may prevent disease onset that is influenced by the genetic and environmental alteration in the expression of susceptibility genes. In contrast, we showed a sparse network in pediatric/adult CD patients, suggesting the broken network contributed to the CD etiology. The network status of susceptibility genes at the single-cell level of monocytes provided novel insight into the etiology.
Collapse
Affiliation(s)
- Hankui Liu
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Genomics, Shenzhen, China
| | - Liping Guan
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Genomics, Shenzhen, China
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Xi Su
- BGI Genomics, Shenzhen, China
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Lijian Zhao
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Genomics, Shenzhen, China
- Hebei Medical University, Shijiazhuang, China
| | - Qing Shu
- Department of Gastroenterology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jianguo Zhang
- Hebei Industrial Technology Research Institute of Genomics in Maternal & Child Health, Clin Lab, BGI Genomics, Shijiazhuang, China
- BGI Research, Shenzhen, China
- Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
24
|
Jia R, Zheng H, Li S, Chen W, Yang Y, Wu H, Chen H, Qin S, Huang S. QingChang-XiaoPi decoction ameliorates intestinal inflammation of ulcerative colitis by regulating the pathogenicity of Th17 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155779. [PMID: 38876011 DOI: 10.1016/j.phymed.2024.155779] [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: 01/31/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND QingChang-XiaoPi Decoction (QCXPY), a Chinese herbal prescription, has been employed in the treatment of ulcerative colitis (UC) in China. However, its molecular mechanism of action in UC remains unclear. PURPOSE To elucidate the therapeutic effects of QCXPY against UC and reveal its mechanism of action. STUDY DESIGN We conducted a single-arm observation to evaluate the clinical efficacy of QCXPY in patients with mild-to-moderate UC. Inclusion and exclusion criteria were established to ensure the eligibility of participants, with a focus on excluding patients with specific conditions or complications that could confound the results. METHODS The expression of inflammatory factors in patients' serum was detected using a Luminex assay. The main components of QCXPY were identified using UHPLC-Q-TOF-MS. Network pharmacology was employed to predict potential therapeutic targets and their mechanisms of action. The efficacy of QCXPY was evaluated using a dextran sulfate sodium (DSS)-induced mouse model. Disease activity index (DAI), histopathological score, cytokine detection by ELISA, T-helper 17 (Th17) cell proportion by flow cytometry, expression of the IL-23/IL-17 axis, and changes in the levels of its downstream effectors were detected by immunohistochemistry, immunofluorescence, and western blotting. RESULTS QCXPY could alleviate the symptoms of diarrhea, abdominal pain, abdominal distension, and purulent stool in patients with mild-to-moderate UC. Moreover, it reduced the expression of IL-6, IL-17, and IL-23 in serum; alleviated DSS-induced experimental colitis in mice; reduced DAI, pathological scores, and the expressions of IL-6, IL-17, and IL-23 in colon tissue; and decreased the proportion of pathogenic Th17 cells and the expression of STAT3 and phospho-STAT3. CONCLUSION This study confirmed for the first time that QCXPY could alleviate intestinal symptoms, reduce the levels of serum inflammatory factors, and improve the quality of life of patients with mild-to-moderate UC. Its mechanism of action may involve reducing the secretion of inflammatory cytokines, moderating the pathogenicity of Th17 cells, and inhibiting STAT3 phosphorylation, thereby alleviating intestinal inflammation in UC.
Collapse
Affiliation(s)
- Rui Jia
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Huan Zheng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China
| | - Siya Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Weihuan Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Yuanming Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Haomeng Wu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China
| | - Haiming Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China
| | - Shumin Qin
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China.
| | - Shaogang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510120, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China; Yang Chunbo academic experience inheritance studio of Guangdong provincial hospital of Chinese Medicine, Guangzhou 510120, PR China.
| |
Collapse
|
25
|
Wang Y, Lai W, Zheng X, Li K, Zhang Y, Pang X, Gao J, Lou Z. Linderae Radix extract attenuates ulcerative colitis by inhibiting the JAK/STAT signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155868. [PMID: 39032278 DOI: 10.1016/j.phymed.2024.155868] [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: 02/07/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Linderae Radix (LR), the dried root of Lindera aggregata (Sims) Kosterm., is a traditional Chinese herbal medicine that has been used for thousands of years for promoting Qi circulation, soothing the liver, and treating diarrhea and dysentery. Previous studies have found that ethanol extract of LR plays an anti-ulcerative colitis (UC) role by regulating Th17/ Treg balance. Water extract is the classic clinical application form of LR, but the effect of water extract of LR (LRWE) on UC and its underlying mechanism is still unclear. PURPOSE Purpose: UC is a gastrointestinal disease characterized by intestinal inflammation, mucosal injury, and fibrosis, and it is one of the high-risk factors for colon cancer. However, there is still a lack of remedies with satisfactory effects. This study aimed to investigate the efficacy and the potential mechanism of LRWE against UC. METHODS LRWE samples were prepared using a reflux extraction method. Colitis in mice was induced by administering 2.5 % DSS water solution to evaluate the therapeutic effect of LRWE by assessing disease activity score, colon length, and fecal morphology. H&E staining, TEM, Masson staining, and AB-PAS staining were applied to observe histopathological changes in the colon tissues. Differentially expressed genes in colon tissues were analyzed by transcriptomics. Cell apoptosis was detected by TUNEL staining. The expression of inflammatory factors such as IL-6 and IL-1β, as well as the expression of p-STAT1, p-JAK2, p-STAT3, Bax, and Bcl-2, were detected by immunofluorescence and immunohistochemistry. The expression of occludin, Bcl-2, Bax, and JAK/STAT signaling pathway-related vital proteins were quantified by Western blot (WB). RESULTS LRWE alleviated body weight loss, colon shortening, DAI scores, pathological changes, and ultrastructural features of colon tissue in mice with colitis. It also inhibited the increase of pro-inflammatory cytokines (such as TNF-α, IL-6, and IL-1β) and increased IL-10 levels. Additionally, it protected the intestinal barrier by upregulating the expression of Occludin and Mucin-2. Mechanistically, LRWE could inhibit the activation of JAK-STAT signaling pathway by reducing the protein expression of p-JAK2, p-STAT3, p-STAT1, Bcl2, and Bax, thus reducing the inflammatory responses and inhibiting cell apoptosis. CONCLUSION LRWE has a protective effect on DSS-induced UC. This effect is related to the inhibition of the JAK-STAT signaling pathway, the improvement of intestinal inflammation, and the reduction of intestinal epithelial cell apoptosis.
Collapse
Affiliation(s)
- Yingying Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China
| | - Weizhi Lai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China
| | - Xunjie Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China
| | - Ke Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China
| | - Yanhua Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China
| | - Xiaojun Pang
- Gastroenterology Department, Tiantai People's Hospital of Zhejiang Province, Zhejiang, Taizhou 317200, PR China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China.
| | - Zhaohuan Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, Hangzhou 310053, PR China; Songyang Institute of Zhejiang Chinese Medical University, Zhejiang, Lishui 323400, PR China.
| |
Collapse
|
26
|
He Y, Shen X, Peng H. Effects and Mechanisms of the Xianhecao-Huanglian Drug Pair on Autophagy-Mediated Intervention in Acute Inflammatory Bowel Disease via the JAK2/STAT3 Pathway. Biol Proced Online 2024; 26:27. [PMID: 39187810 PMCID: PMC11346250 DOI: 10.1186/s12575-024-00242-5] [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: 02/05/2024] [Accepted: 05/09/2024] [Indexed: 08/28/2024] Open
Abstract
To explore the effects and mechanisms of the Xianhecao-Huanglian drug pair on autophagy-mediated intervention in acute inflammatory bowel disease (IBD) via the JAK2/STAT3 pathway. The study examined the underlying mechanisms of action of Xianhecao (APL) and Huanglian (CR) using a mouse model of dextran sodium sulfate (DSS)-induced acute inflammatory bowel disease (IBD) and in an in vitro model of IBD induced by lipopolysaccharide (LPS). The assessment of the therapeutic efficacy of the Xianhecao-Huanglian drug combination in a mouse model of IBD caused by DSS included the following parameters: Assessment of weight loss or gain. Measurement of the disease activity index (DAI). Assessment of histological damage. Determination of organ index. Measurement of colon length. Ascertain the levels of inflammatory cytokines in the intestinal tissues and serum of mice. Immunohistochemistry (IHC) for the measurement of tight junction protein concentrations in the colon mucosa, including ZO-1, claudin-1, and occludin. Measurement of mucin levels, specifically Mucin 2 (Muc2). Hematoxylin and eosin (HE) staining for the observation of histopathological alterations in colonic tissues. Examining the effect on goblet cells using periodic acid-Schiff (PAS) labeling. Application of Western blot and immunofluorescence techniques for the detection of autophagy-related markers in colonic tissues and proteins associated with the JAK2/STAT3 pathway. A cell inflammation model of IBD was induced through LPS stimulation, and a serum containing the Xianhecao-Huanglian drug pair (referred to as ACHP-DS) was formulated. Cell viability, anti-proinflammatory cytokines, tight junction proteins, mucins, autophagy-related markers, and the JAK2/STAT3 signaling pathway were assessed. The Xianhecao-Huanglian drug pair significantly ameliorated the symptoms and survival quality of acute IBD mice, reducing the disease activity index score, raising MUC2 secretion and tight junction protein expression to improve the integrity of the intestinal barrier, and preserving goblet cell function; thus, protecting the intestines. It effectively restrained triggering the signaling pathway that involves JAK2 and STAT3, leading to the suppression of inflammation and amelioration of colonic inflammation damage. Additionally, it induced autophagy in mouse colonic tissues.The in vitro experiments demonstrated that the Xianhecao-Huanglian drug combination enhanced the viability of LOVO and NCM460 cells when exposed to LPS stimulation. Furthermore, it suppressed the production of inflammatory cytokines such as IL-6, IL-1β, as well as TNF-α, whilst increasing the production of IL-10, ZO-1, along with MUC2. These effects collectively led to the alleviation of inflammation and the restoration of mucosal integrity. The results were consistent with what was shown in the in vivo trial. Moreover, the medication demonstrated effectiveness in reducing JAK2 along with STAT3 phosphorylation levels in the LPS-induced inflammatory model of IBD cells. The intervention with either the Xianhecao-Huanglian drug combination-containing serum or the JAK2/STAT3 pathway inhibitor AG490 reversed the pro-inflammatory effects and increased autophagy levels in the LPS-stimulated cells. The Xianhecao-Huanglian drug combination modulates the JAK2/STAT3 pathway, leading to the induction of autophagy, which serves as an intervention for IBD.
Collapse
Affiliation(s)
- Yaping He
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| | - Xinling Shen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Haiyan Peng
- The First Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
27
|
Bao X, Tang Y, Lv Y, Fu S, Yang L, Chen Y, Zhou M, Zhu B, Ding Z, Zhou F. Tetrastigma hemsleyanum polysaccharide ameliorated ulcerative colitis by remodeling intestinal mucosal barrier function via regulating the SOCS1/JAK2/STAT3 pathway. Int Immunopharmacol 2024; 137:112404. [PMID: 38851163 DOI: 10.1016/j.intimp.2024.112404] [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: 01/19/2024] [Revised: 05/21/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Ulcerative colitis (UC) is characterized by a chronic and protracted course and often leads to a poor prognosis. Patients with this condition often experience postoperative complications, further complicating the management of their condition. Tetrastigma hemsleyanum polysaccharide (THP) has demonstrated considerable potential as a treatment for inflammatory bowel disease. However, its underlying mechanism in the treatment of UC remains unclear. This study systematically and comprehensively investigated the effects of THP on dextran sulfate-induced UC mice and illustrated its specific mechanism of action. The colon and spleen in UC mice were restored after THP treatment. The levels of key markers, such as secretory immunoglobulin A, β-defensin, and mucin-2 were increased, collagen deposition and epithelial cell apoptosis were decreased. Notably, THP administration led to increased levels of Ki67 and tight junction proteins in colon tissue and reduced colon tissue permeability. THP contributed to the restored balance of intestinal flora. Furthermore, THP downregulated the expressions of the proinflammatory cytokines interleukin (IL)-6, tumor necrosis factor (TNF)-α, and IL-17 and promoted those of the regulatory factors forkhead box protein P3. It also exerted anti-inflammatory effects by promoting suppressor of cytokine signaling (SOCS1) expression and inhibiting the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Our results demonstrated that THP had an efficacy comparable to that of JAK inhibitor in treating UC. In addition, THP might play a role in UC therapy through modulation of the SOCS1/JAK2/STAT3 signaling pathway and remodeling of the intestinal mucosal barrier.
Collapse
Affiliation(s)
- Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Youying Tang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yishan Lv
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Liu Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yuchi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Mingyuan Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Bingqi Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
28
|
Baars MJ, Floor E, Sinha N, ter Linde JJ, van Dam S, Amini M, Nijman IJ, ten Hove JR, Drylewicz J, Offerhaus GA, Laclé MM, Oldenburg B, Vercoulen Y. Multiplex spatial omics reveals changes in immune-epithelial crosstalk during inflammation and dysplasia development in chronic IBD patients. iScience 2024; 27:110550. [PMID: 39165839 PMCID: PMC11334790 DOI: 10.1016/j.isci.2024.110550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/16/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Patients with long-standing inflammatory bowel disease (IBD) face an increased risk of developing colitis-associated cancer (CAC). Although IBD-induced prolonged inflammation seems to be involved in CAC pathogenesis, the specific molecular changes that contribute remain unknown. Here, we applied digital spatial RNA profiling, RNAscope, and imaging mass cytometry to examine paired uninflamed, inflamed, and early dysplastic mucosa of patients with IBD. We observed robust type 3 (IL-17) responses during inflammation, accompanied by elevated JAK-STAT signaling and phosphorylated STAT3 (P-STAT3) levels, with both inflamed and dysplastic mucosa displaying immune cell activation. Higher stromal P-STAT3 was detected in uninflamed and inflamed mucosa of patients who eventually developed dysplasia. CD8a+ T cells did not infiltrate inflamed or dysplastic epithelial regions in these patients, while control patients showed elevated CD8a in inflamed mucosa. Our study reveals distinct inflammatory patterns throughout CAC development, marked by an activated IL-17 pathway, engaged STAT3, and diminished cytotoxic T cell infiltration.
Collapse
Affiliation(s)
- Matthijs J.D. Baars
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
| | - Evelien Floor
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX, Utrecht 3584, the Netherlands
| | - Neeraj Sinha
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
| | - José J.M. ter Linde
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX, Utrecht 3584, the Netherlands
| | - Stephanie van Dam
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Mojtaba Amini
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
- UCyTOF, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
| | - Isaäc J. Nijman
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
- USEQ, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
| | - Joren R. ten Hove
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX, Utrecht 3584, the Netherlands
| | - Julia Drylewicz
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, EA, Utrecht 3584, the Netherlands
| | - G.Johan A. Offerhaus
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX, Utrecht 3584, the Netherlands
| | - Miangela M. Laclé
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX, Utrecht 3584, the Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, CX, Utrecht 3584, the Netherlands
| | - Yvonne Vercoulen
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
- UCyTOF, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, CX, Utrecht 3584, the Netherlands
| |
Collapse
|
29
|
Bahman F, Choudhry K, Al-Rashed F, Al-Mulla F, Sindhu S, Ahmad R. Aryl hydrocarbon receptor: current perspectives on key signaling partners and immunoregulatory role in inflammatory diseases. Front Immunol 2024; 15:1421346. [PMID: 39211042 PMCID: PMC11358079 DOI: 10.3389/fimmu.2024.1421346] [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: 04/29/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a versatile environmental sensor and transcription factor found throughout the body, responding to a wide range of small molecules originating from the environment, our diets, host microbiomes, and internal metabolic processes. Increasing evidence highlights AhR's role as a critical regulator of numerous biological functions, such as cellular differentiation, immune response, metabolism, and even tumor formation. Typically located in the cytoplasm, AhR moves to the nucleus upon activation by an agonist where it partners with either the aryl hydrocarbon receptor nuclear translocator (ARNT) or hypoxia-inducible factor 1β (HIF-1β). This complex then interacts with xenobiotic response elements (XREs) to control the expression of key genes. AhR is notably present in various crucial immune cells, and recent research underscores its significant impact on both innate and adaptive immunity. This review delves into the latest insights on AhR's structure, activating ligands, and its multifaceted roles. We explore the sophisticated molecular pathways through which AhR influences immune and lymphoid cells, emphasizing its emerging importance in managing inflammatory diseases. Furthermore, we discuss the exciting potential of developing targeted therapies that modulate AhR activity, opening new avenues for medical intervention in immune-related conditions.
Collapse
Affiliation(s)
- Fatemah Bahman
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Khubaib Choudhry
- Department of Human Biology, University of Toronto, Toronto, ON, Canada
| | - Fatema Al-Rashed
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fahd Al-Mulla
- Department of Translational Research, Dasman Diabetes Institute, Dasman, Kuwait
| | - Sardar Sindhu
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
- Animal & Imaging Core Facilities, Dasman Diabetes Institute, Dasman, Kuwait
| | - Rasheed Ahmad
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| |
Collapse
|
30
|
Braidotti S, Granzotto M, Curci D, Faganel Kotnik B, Maximova N. Advancing Allogeneic Hematopoietic Stem Cell Transplantation Outcomes through Immunotherapy: A Comprehensive Review of Optimizing Non-CAR Donor T-Lymphocyte Infusion Strategies. Biomedicines 2024; 12:1853. [PMID: 39200317 PMCID: PMC11351482 DOI: 10.3390/biomedicines12081853] [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: 07/04/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Optimized use of prophylactic or therapeutic donor lymphocyte infusions (DLI) is aimed at improving clinical outcomes in patients with malignant and non-malignant hematological diseases who have undergone allogeneic hematopoietic stem cell transplantation (allo-HSCT). Memory T-lymphocytes (CD45RA-/CD45RO+) play a crucial role in immune reconstitution post-HSCT. The infusion of memory T cells is proven to be safe and effective in improving outcomes due to the enhanced reconstitution of immunity and increased protection against viremia, without exacerbating graft-versus-host disease (GVHD) risks. Studies indicate their persistence and efficacy in combating viral pathogens, suggesting a viable therapeutic avenue for patients. Conversely, using virus-specific T cells for viremia control presents challenges, such as regulatory hurdles, cost, and production time compared to CD45RA-memory T lymphocytes. Additionally, the modulation of regulatory T cells (Tregs) for therapeutic use has become an important area of investigation in GVHD, playing a pivotal role in immune tolerance modulation, potentially mitigating GVHD and reducing pharmacological immunosuppression requirements. Finally, donor T cell-mediated graft-versus-leukemia immune responses hold promise in curbing relapse rates post-HSCT, providing a multifaceted approach to therapeutic intervention in high-risk disease scenarios. This comprehensive review underscores the multifaceted roles of T lymphocytes in HSCT outcomes and identifies avenues for further research and clinical application.
Collapse
Affiliation(s)
- Stefania Braidotti
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Marilena Granzotto
- Azienda Sanitaria Universitaria Giuliano Isontina (ASU GI), 34125 Trieste, Italy;
| | - Debora Curci
- Advanced Translational Diagnostic Laboratory, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| | - Barbara Faganel Kotnik
- Department of Hematology and Oncology, University Children’s Hospital, 1000 Ljubljana, Slovenia;
| | - Natalia Maximova
- Department of Pediatrics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, 34137 Trieste, Italy;
| |
Collapse
|
31
|
van Gennep S, Fung ICN, de Jong DC, Ramkisoen RK, Clasquin E, de Jong J, de Vries LCS, de Jonge WJ, Gecse KB, Löwenberg M, Woolcott JC, Mookhoek A, D’Haens GR. Histological Outcomes and JAK-STAT Signalling in Ulcerative Colitis Patients Treated with Tofacitinib. J Crohns Colitis 2024; 18:1283-1291. [PMID: 38506097 PMCID: PMC11324337 DOI: 10.1093/ecco-jcc/jjae031] [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: 10/24/2023] [Revised: 01/26/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND AND AIMS Histological outcomes and JAK-STAT signalling were assessed in a prospective ulcerative colitis [UC] patient cohort after 8 weeks treatment with tofacitinib, an oral Janus kinase [JAK] inhibitor. METHODS Forty UC patients received tofacitinib 10 mg twice daily for 8 weeks. Treatment response was defined as histo-endoscopic mucosal improvement [HEMI]. Histological remission was defined as a Robarts Histopathology Index [RHI] ≤3 points and histological response as 50% decrease in RHI. Mucosal expression of JAK1-3, tyrosine kinase 2 [TYK2], and total signal transducer and activator of transcription [STAT] 1-6 were assessed using immunohistochemistry [IHC]. RESULTS At baseline, the median RHI was 14 (interquartile range [IQR] 10-19). Of 40 [65%] patients, 26 had severe endoscopic disease [endoscopic Mayo score 3] and 31/40 [78%] failed prior anti-tumour necrosis factor [anti-TNF] treatment. At Week 8, 15 patients [38%] had HEMI, 23 patients [58%] histological remission, and 34 [85%] histological response. RHI decreased by a median of 14 points [IQR 9-21] in responders [p <0.001] and by 6 points [IQR 0-13] in non-responders [p = 0.002]. STAT1, STAT3, and STAT5 expression levels decreased significantly in the whole cohort. Responders had lower Week 8 STAT1 expression levels compared with non-responders [0.2%, IQR 0.1-2.8 vs 4.3%, IQR 1.2-11.9, p = 0.001], suggesting more profound STAT1 blockade. A trend of higher baseline JAK2 expression was observed in tofacitinib non-responders [2.7%, IQR 0.1-7.7] compared with responders [0.4%, IQR 0.1-2.1]. CONCLUSIONS Tofacitinib treatment resulted in histological improvement in the majority of UC patients and in a substantial decrease of STAT1, STAT3, and STAT5 expression. HEMI was associated with more profound suppression of STAT1.
Collapse
Affiliation(s)
- Sara van Gennep
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Ivan C N Fung
- Amsterdam UMC, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Djuna C de Jong
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Rishand K Ramkisoen
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Esmé Clasquin
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Jitteke de Jong
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Leonie C S de Vries
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Wouter J de Jonge
- Amsterdam UMC, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands
| | - Krisztina B Gecse
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| | | | - Aart Mookhoek
- University of Bern, Department of Pathology, Institute of Tissue Medicine and Pathology, Bern, Switzerland
| | - Geert R D’Haens
- Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Joustra V, Li Yim AYF, van Gennep S, Hageman I, de Waard T, Levin E, Lauffer P, de Jonge W, Henneman P, Löwenberg M, D’Haens G. Peripheral Blood DNA Methylation Signatures and Response to Tofacitinib in Moderate-to-severe Ulcerative Colitis. J Crohns Colitis 2024; 18:1179-1189. [PMID: 37526299 PMCID: PMC11324342 DOI: 10.1093/ecco-jcc/jjad129] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/22/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
INTRODUCTION Predictive biomarkers for treatment efficacy of ulcerative colitis [UC] treatments are lacking. Here, we performed a longitudinal study investigating the association and potential predictive power of genome-wide peripheral blood [PB] DNA methylation signatures and response to tofacitinib treatment in UC. METHODS We recruited moderate-to-severe UC patients starting tofacitinib treatment, and measured PB DNA methylation profiles at baseline [T1], after 8 weeks [T2], and in a subset [n = 8] after a median of 20 weeks [T3] using the Illumina Infinium HumanMethylation EPIC BeadChip. After 8 weeks, we distinguished responders [R] from non-responders [NR] based on a centrally read endoscopic response [decrease in endoscopic Mayo score ≥1 or Ulcerative Colitis Endoscopic Index of Severity ≥2] combined with corticosteroid-free clinical and/or biochemical response. T1 PB samples were used for biomarker identification, and T2 and publicly available intraclass correlation [ICC] data were used for stability analyses. RNA-sequencing was performed to understand the downstream effects of the predictor CpG loci. RESULTS In total, 16 R and 15 NR patients, with a median disease duration of 7 [4-12] years and overall comparable patient characteristics at baseline, were analysed. We identified a panel of 53 differentially methylated positions [DMPs] associated with response to tofacitinib [AUROC 0.74]. Most DMPs [77%] demonstrated both short- and long-term hyperstability [ICC ≥0.90], irrespective of inflammatory status. Gene expression analysis showed lower FGFR2 [pBH = 0.011] and LRPAP1 [pBH = 0.020], and higher OR2L13 [pBH = 0.016] expression at T1 in R compared with NR. CONCLUSION Our observations demonstrate the utility of genome-wide PB DNA methylation signatures to predict response to tofacitinib.
Collapse
Affiliation(s)
- Vincent Joustra
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Andrew Y F Li Yim
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Genome Diagnostics Laboratory, Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sara van Gennep
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ishtu Hageman
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Peter Lauffer
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Wouter de Jonge
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Peter Henneman
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Genome Diagnostics Laboratory, Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Esai Selvan M, Nathan DI, Guisado D, Collatuzzo G, Iruvanti S, Boffetta P, Mascarenhas J, Hoffman R, Cohen LJ, Marcellino BK, Gümüş ZH. Clonal Hematopoiesis of Indeterminate Potential in Crohn's Disease and Ulcerative Colitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.06.24311497. [PMID: 39148820 PMCID: PMC11326358 DOI: 10.1101/2024.08.06.24311497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is the presence of somatic mutations in myeloid and lymphoid malignancy genes in the blood cells of individuals without a hematologic malignancy. Inflammation is hypothesized to be a key mediator in the progression of CHIP to hematologic malignancy and patients with CHIP have a high prevalence of inflammatory diseases. This study aimed to identify the prevalence and characteristics of CHIP in patients with inflammatory bowel disease (IBD). We analyzed whole exome sequencing data from 587 Crohn's disease (CD), 441 ulcerative colitis (UC), and 293 non-IBD controls to assess CHIP prevalence and used logistic regression to study associations with clinical outcomes. Older UC patients (age>45) harbored increased myeloid-CHIP mutations compared to younger patients (age≤45) (p=0.01). Lymphoid-CHIP was more prevalent in older IBD patients (p=0.007). Young CD patients were found to have myeloid-CHIP with high-risk features. IBD patients with CHIP exhibited unique mutational profiles compared to controls. Steroid use was associated with increased CHIP (p=0.05), while anti-TNF therapy was associated with decreased myeloid-CHIP (p=0.03). Pathway enrichment analyses indicated overlap between CHIP genes, IBD phenotypes, and inflammatory pathways. Our findings underscore a connection between IBD and CHIP pathophysiology. Patients with IBD and CHIP had unique risk profiles especially among older UC patients and younger CD patients. These findings suggest distinct evolutionary pathways for CHIP in IBD and necessitate awareness among IBD providers and hematologists to identify patients potentially at risk for CHIP-related complications including malignancy, cardiovascular disease and acceleration of their inflammatory disease.
Collapse
Affiliation(s)
- Myvizhi Esai Selvan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel I Nathan
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniela Guisado
- Division of Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Stony Brook Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, Stony Brook, NY, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Hoffman
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Louis J Cohen
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bridget K Marcellino
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
34
|
Shah B, Solanki N. Aegeline attenuates TNBS-induced colitis by suppressing the NFƙB-mediated NLRP3 inflammasome pathway in mice. Inflammopharmacology 2024; 32:2589-2599. [PMID: 38767762 DOI: 10.1007/s10787-024-01493-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
A chronic inflammatory condition of the intestine, ulcerative colitis (UC), is challenging to successfully manage once diagnosed. Currently, available medical therapies for UC exhibit minimal efficacy with unacceptable side effects, while inventive biological agents are expensive and yet not well accepted by patients. Discovering more effective and safer treatments to treat UC is therefore essential. One of the primary alkaloids found in Aegle marmelos, aegeline, has anti-inflammatory and antioxidant properties as well as being able to suppress several pro-inflammatory cytokines responsible for inflammation. The study aimed to investigate the effectiveness of aegeline in alleviating 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis through the NFƙB-mediated NLRP3 inflammasome pathway. Mice were randomly allocated into six groups, Normal control (NC), Model control (MC-TNBS, 2,4,6-trinitrobenzene sulfonic acid), STD (TNBS + sulfasalazine 100 mg/kg), AG1, AG2, and AG3 (TNBS + aegeline 5, 10, 20 mg/kg) respectively. Physical parameters such as a change in body weight, stool consistency, rectal bleeding, colon length, myeloperoxidase (MPO) levels and nitric oxide (NO) levels, and disease activity index (DAI) were assessed and supporting gene expression studies of various pro-inflammatory cytokines and enzymes were evaluated and histopathological changes observed. Administration of aegeline (10, 20 mg/kg) was found to be effective in colon protection by lowering the disease activity score and myeloperoxidase level and improving other physical parameters. Aegeline in high dose significantly downregulated the gene expression of NFƙB, iNOS, COX-2, NLRP3, IL-1β, and IL-18, conferring great anti-inflammatory potential. Suggestive of the findings, aegeline reduced the damage to the colon by downregulating transcriptional genes and enzymes leading to inflammation and mitigated TNBS-induced colitis probably through the NFƙB-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Bhagyabhumi Shah
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa, 388421, Gujarat, India.
| | - Nilay Solanki
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa, 388421, Gujarat, India.
| |
Collapse
|
35
|
Jeong YJ, Lee HR, Park SA, Lee JW, Kim LK, Kim HJ, Seo JH, Heo TH. A derivative of 3-(1,3-diarylallylidene)oxindoles inhibits dextran sulfate sodium-induced colitis in mice. Pharmacol Rep 2024; 76:851-862. [PMID: 38916850 PMCID: PMC11294400 DOI: 10.1007/s43440-024-00616-2] [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: 02/12/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND IA-0130 is a derivative of 3-(1,3-diarylallylidene)oxindoles, which is a selective estrogen receptor modulator (SERM). A previous study demonstrated that SERM exhibits anti-inflammatory effects on colitis by promoting the anti-inflammatory phenotype of monocytes in murine colitis. However, the therapeutic effects of oxindole on colitis remain unknown. Therefore, we evaluated the efficacy of IA-0130 on dextran sulfate sodium (DSS)-induced mouse colitis. METHODS The DSS-induced colitis mouse model was established by administration of 2.5% DSS for 5 days. Mice were orally administered with IA-0130 (0.01 mg/kg or 0.1 mg/kg) or cyclosporin A (CsA; 30 mg/kg). Body weight, disease activity index score and colon length of mice were calculated and histological features of mouse colonic tissues were analyzed using hematoxylin and eosin staining. The expression of inflammatory cytokines and tight junction (TJ) proteins were analyzed using quantitative real-time PCR and enzyme-linked immunosorbent assay. The expression of interleukin-6 (IL-6) signaling molecules in colonic tissues were investigated using Western blotting and immunohistochemistry (IHC). RESULTS IA-0130 (0.1 mg/kg) and CsA (30 mg/kg) prevented colitis symptom, including weight loss, bleeding, colon shortening, and expression of pro-inflammatory cytokines in colon tissues. IA-0130 treatment regulated the mouse intestinal barrier permeability and inhibited abnormal TJ protein expression. IA-0130 down-regulated IL-6 expression and prevented the phosphorylation of signaling molecules in colonic tissues. CONCLUSIONS This study demonstrated that IA-0130 suppressed colitis progression by inhibiting the gp130 signaling pathway and expression of pro-inflammatory cytokines, and maintaining TJ integrity.
Collapse
Affiliation(s)
- Young-Jin Jeong
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Hae-Ri Lee
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Sun-Ae Park
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Joong-Woon Lee
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Lee Kyung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Hee Jung Kim
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea
| | - Jae Hong Seo
- Laboratory of Pharmaceutical Manufacturing Chemistry, Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, 43 Jibong-Ro, Bucheon‑si, Gyeonggi‑do, 14662, Republic of Korea
| | - Tae-Hwe Heo
- Laboratory of Pharmacoimmunology, Integrated Research Institute of Pharmaceutical Sciences, BK21 FOUR Team for Advanced Program for SmartPharma Leaders, College of Pharmacy, The Catholic University of Korea, NP512, Hall of Cardinal Jin-Suk Cheong, 43 Jibong-Ro, Bucheon-Si, Gyeonggi‑do, 14662, Republic of Korea.
| |
Collapse
|
36
|
Zhang Y, Zhang C, He J, Lai G, Li W, Zeng H, Zhong X, Xie B. Comprehensive analysis of single cell and bulk RNA sequencing reveals the heterogeneity of melanoma tumor microenvironment and predicts the response of immunotherapy. Inflamm Res 2024; 73:1393-1409. [PMID: 38896289 DOI: 10.1007/s00011-024-01905-5] [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: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Tumor microenvironment (TME) heterogeneity is an important factor affecting the treatment response of immune checkpoint inhibitors (ICI). However, the TME heterogeneity of melanoma is still widely characterized. METHODS We downloaded the single-cell sequencing data sets of two melanoma patients from the GEO database, and used the "Scissor" algorithm and the "BayesPrism" algorithm to comprehensively analyze the characteristics of microenvironment cells based on single-cell and bulk RNA-seq data. The prediction model of immunotherapy response was constructed by machine learning and verified in three cohorts of GEO database. RESULTS We identified seven cell types. In the Scissor+ subtype cell population, the top three were T cells, B cells and melanoma cells. In the Scissor- subtype, there are more macrophages. By quantifying the characteristics of TME, significant differences in B cells between responders and non-responders were observed. The higher the proportion of B cells, the better the prognosis. At the same time, macrophages in the non-responsive group increased significantly. Finally, nine gene features for predicting ICI response were constructed, and their predictive performance was superior in three external validation groups. CONCLUSION Our study revealed the heterogeneity of melanoma TME and found a new predictive biomarker, which provided theoretical support and new insights for precise immunotherapy of melanoma patients.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Cong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Jing He
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Wenlong Li
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Haijiao Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Xiaoni Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China.
| | - Biao Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
37
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Campen M, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances Th17 Responses and Promotes Airway Neutrophilia. Am J Respir Cell Mol Biol 2024; 71:169-181. [PMID: 38593442 PMCID: PMC11299091 DOI: 10.1165/rcmb.2023-0424oc] [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: 12/01/2023] [Accepted: 04/09/2024] [Indexed: 04/11/2024] Open
Abstract
Heightened unfolded protein responses (UPRs) are associated with the risk for asthma, including severe asthma. Treatment-refractory severe asthma manifests a neutrophilic phenotype with T helper (Th)17 responses. However, how UPRs participate in the deregulation of Th17 cells leading to neutrophilic asthma remains elusive. This study found that the UPR sensor IRE1 is induced in the murine lung with fungal asthma and is highly expressed in Th17 cells relative to naive CD4+ T cells. Cytokine (e.g., IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by both human and mouse Th17 cells. Ern1 (encoding IRE1) deficiency decreases the expression of endoplasmic reticulum stress factors and impairs the differentiation and cytokine secretion of Th17 cells. Genetic ablation of Ern1 leads to alleviated Th17 responses and airway neutrophilia in a fungal airway inflammation model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances Th17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPR-mediated secretory function of Th17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in Th17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology and
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | | - Ruoning Wang
- Department of Molecular Genetics and Microbiology and
| | | | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas; and
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology and
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | | | - Meilian Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | |
Collapse
|
38
|
Olivera PA, Dignass A, Dubinsky MC, Peretto G, Kotze PG, Dotan I, Kobayashi T, Ghosh S, Magro F, Faria-Neto JR, Siegmund B, Danese S, Peyrin-Biroulet L. Preventing and managing cardiovascular events in patients with inflammatory bowel diseases treated with small-molecule drugs, an international Delphi consensus. Dig Liver Dis 2024; 56:1270-1280. [PMID: 38584033 DOI: 10.1016/j.dld.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/08/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024]
Abstract
Janus kinase (JAK) inhibitors and sphingosine 1 phosphate (S1P) receptor modulators are small molecule drugs (SMDs) approved for IBD treatment. Their use in clinical practice might be limited due to cardiovascular concerns. We aimed to provide guidance on risk assessment, monitoring, and management strategies, aiming to minimize potential cardiovascular risks of SMDs and to facilitate an adequate shared decision-making. A systematic literature search was conducted, and proposed statements were prepared. A virtual consensus meeting was held, in which eleven IBD physicians and two cardiovascular specialists from ten countries attended. Proposed statements were voted upon in an anonymous manner. Agreement was defined as at least 75 % of participants voting as 'agree' with each statement. Consensus was reached for eighteen statements. Available evidence does not show a higher risk of cardiovascular events with JAK inhibitors in the overall IBD population, although it might be increased in patients with an unfavorable cardiovascular profile. S1P receptor modulators may be associated with a risk of bradycardia, atrioventricular blocks, and hypertension. Cardiovascular risk stratification should be done before initiation of SMDs. Although the risk of cardiovascular events in patients with IBD on SMDs appears to be low overall, caution should still be taken in certain scenarios.
Collapse
Affiliation(s)
- Pablo A Olivera
- IBD Unit, Gastroenterology Section, Department of Internal Medicine, Centro de Educación Médica e Investigación Clínica (CEMIC), Buenos Aires, Argentina; Zane Cohen Centre for Digestive Diseases, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Axel Dignass
- Department of Medicine I, Agaplesion Markus Hospital, Goethe-University, Frankfurt Am Main, Germany
| | - Marla C Dubinsky
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Giovanni Peretto
- Myocarditis Disease Unit, Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, Milan, Italy; School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Paulo G Kotze
- IBD outpatient clinics, Colorectal Surgery Unit, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Iris Dotan
- Sackler Faculty of Medicine, Tel Aviv University, Israel; Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
| | - Taku Kobayashi
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, Tokyo, Japan
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Fernando Magro
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Jose Rocha Faria-Neto
- School of Medicine, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Britta Siegmund
- Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milano, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; INSERM, NGERE, University of Lorraine, F-54000 Nancy, France; INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; Groupe Hospitalier Privé Ambroise Paré - Hartmann, Paris IBD center, 92200 Neuilly sur Seine, France; Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
39
|
Li B, Hu P, Liang H, Zhao X, Zhang A, Xu Y, Zhang B, Zhang J. Evaluating the causal effect of circulating proteome on the risk of inflammatory bowel disease-related traits using Mendelian randomization. Front Immunol 2024; 15:1434369. [PMID: 39144148 PMCID: PMC11321985 DOI: 10.3389/fimmu.2024.1434369] [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: 05/17/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Objective This study sought to identify circulating proteins causally linked to Inflammatory Bowel Disease (IBD) traits through a Mendelian Randomization (MR) analytical framework. Methods Using a large-scale, two-sample MR approach, we estimated the genetic links of numerous plasma proteins with IBD and its subtypes, leveraging information from the Inflammatory Bowel Disease Genetics Consortium. To assess the robustness of MR findings, methods like Bayesian colocalization, and Steiger filtering analysis, evaluation of protein-altering variants. Further insights into IBD's underlying mechanisms and therapeutic targets were gleaned from single-cell sequencing analyses, protein-protein interaction assessments, pathway enrichment analyses, and evaluation of drug targets. Results By cis-only MR analysis, we identified 83 protein-phenotype associations involving 27 different proteins associated with at least one IBD subtype. Among these proteins, DAG1, IL10, IL12B, IL23R, MST1, STAT3 and TNFRSF6B showed overlapping positive or negative associations in all IBD phenotypes. Extending to cis + trans MR analysis, we further identified 117 protein-feature associations, including 44 unique proteins, most of which were not detected in the cis-only analysis. In addition, by performing co-localization analysis and Steiger filtering analysis on the prioritized associations, we further confirmed the causal relationship between these proteins and the IBD phenotype and verified the exact causal direction from the protein to the IBD-related feature. Conclusion MR analysis facilitated the identification of numerous circulating proteins associated with IBD traits, unveiling protein-mediated mechanisms and promising therapeutic targets.
Collapse
Affiliation(s)
- Beining Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ping Hu
- Department of Orthopedic, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyan Liang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xingliang Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Aiting Zhang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yingchong Xu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bin Zhang
- Department of Orthopedic, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Zhang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| |
Collapse
|
40
|
Liu Y, Song X, Xiang L, Tan W, Zou M, Guo H. Rapid Remission With Upadacitinib in a Child With Refractory Crohn's Disease and ATM Mutation: A Case Report. CURRENT THERAPEUTIC RESEARCH 2024; 101:100756. [PMID: 39257480 PMCID: PMC11385748 DOI: 10.1016/j.curtheres.2024.100756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/20/2024] [Indexed: 09/12/2024]
Abstract
Managing pediatric Crohn's disease (PCD) presents challenges due to severe complications and higher biologic therapy needs. Transitioning from anti-tumor necrosis factor agents to off-label therapies adds complexity. Although upadacitinib has demonstrated efficacy and tolerability in adult inflammatory bowel disease and pediatric atopic dermatitis, there are limited data for its application in PCD. This case report delineates successful remission with upadacitinib in a child with CD refractory to infliximab, ustekinumab, adalimumab, thalidomide, and prednisone. Notably, the patient carried an ataxia telangiectasia mutated (ATM) gene mutation. These findings provide valuable evidence for PCD management and highlight the potential benefits of upadacitinib in this population.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - XiaoMei Song
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - LingYa Xiang
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Wei Tan
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Zou
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Hong Guo
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
41
|
Liu X, Tang Y, Luo Y, Gao Y, He L. Role and mechanism of specialized pro-resolving mediators in obesity-associated insulin resistance. Lipids Health Dis 2024; 23:234. [PMID: 39080624 PMCID: PMC11290132 DOI: 10.1186/s12944-024-02207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/07/2024] [Indexed: 08/02/2024] Open
Abstract
With the changing times, obesity has become a characteristic epidemic in the context of the current era. Insulin resistance (IR) is most commonly caused by obesity, and IR is a common basis of the pathogenesis of many diseases such as cardiovascular disease, nonalcoholic fatty liver disease, and type 2 diabetes, which seriously threaten human life, as well as health. A major pathogenetic mechanism of obesity-associated IR has been found to be chronic low-grade inflammation in adipose tissue. Specialized pro-resolving mediators (SPMs) are novel lipid mediators that both function as "stop signals" for inflammatory reaction and promote inflammation to subside. In this article, we summarize the pathogenesis of obesity-associated IR and its treatments and outline the classification and biosynthesis of SPMs and their mechanisms and roles in the treatment of obesity-associated IR in order to explore the potential of SPMs for treating metabolic diseases linked with obesity-associated IR.
Collapse
Affiliation(s)
- Xinru Liu
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Tang
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanyuan Luo
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongxiang Gao
- College of International Education, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lisha He
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
42
|
Esmaeili N, Bakheet A, Tse W, Liu S, Han X. Interaction of the intestinal cytokines-JAKs-STAT3 and 5 axes with RNA N6-methyladenosine to promote chronic inflammation-induced colorectal cancer. Front Oncol 2024; 14:1352845. [PMID: 39136000 PMCID: PMC11317299 DOI: 10.3389/fonc.2024.1352845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 06/25/2024] [Indexed: 08/15/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers, with a high mortality rate worldwide. Mounting evidence indicates that mRNA modifications are crucial in RNA metabolism, transcription, processing, splicing, degradation, and translation. Studies show that N6-methyladenosine (m6A) is mammalians' most common epi-transcriptomic modification. It has been demonstrated that m6A is involved in cancer formation, progression, invasion, and metastasis, suggesting it could be a potential biomarker for CRC diagnosis and developing therapeutics. Cytokines, growth factors, and hormones function in JAK/STAT3/5 signaling pathway, and they could regulate the intestinal response to infection, inflammation, and tumorigenesis. Reports show that the JAK/STAT3/5 pathway is involved in CRC development. However, the underlying mechanism is still unclear. Signal Transducer and Activator of Transcription 3/5 (STAT3, STAT5) can act as oncogenes or tumor suppressors in the context of tissue types. Also, epigenetic modifications and mutations could alter the balance between pro-oncogenic and tumor suppressor activities of the STAT3/5 signaling pathway. Thus, exploring the interaction of cytokines-JAKs-STAT3 and/or STAT5 with mRNA m6A is of great interest. This review provides a comprehensive overview of the characteristics and functions of m6A and JAKs-STAT3/5 and their relationship with gastrointestinal (GI) cancers.
Collapse
Affiliation(s)
- Nardana Esmaeili
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - Ahmed Bakheet
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - William Tse
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - Shujun Liu
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
| | - Xiaonan Han
- Division of Hematology and Oncology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center (MHMC), Case Western Reserve University (CWRU) School of Medicine, Cleveland, OH, United States
- Cancer Genomics and Epigenomics Program, Case Comprehensive Cancer Center, Case Western Reserve University (CWRU), Cleveland, OH, United States
| |
Collapse
|
43
|
Tian Z, Zhao Q, Teng X. Anti-IL23/12 agents and JAK inhibitors for inflammatory bowel disease. Front Immunol 2024; 15:1393463. [PMID: 39086483 PMCID: PMC11288814 DOI: 10.3389/fimmu.2024.1393463] [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: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
IBD (inflammatory bowel disease) is a chronic inflammatory disease of the gastrointestinal tract with increasing incidence worldwide. Multiple factors, such as genetic background, environmental and luminal factors, and mucosal immune dysregulation, have been implicated in the cause of IBD, although the cause of the disease remains unknown. IL-12 and IL-23 and their downstream signaling pathways participate in the pathogenesis of inflammatory bowel disease. Early and aggressive treatment with biologic therapies or novel small molecules is needed to decrease complications and the need for hospitalization and surgery. The landscape of inflammatory bowel disease (IBD) treatment has tremendously improved with the development of biologics and small molecule drugs. Several novel biologics and small molecule drugs targeting IL-12 and IL-23 and their downstream targets have shown positive efficacy and safety data in clinical trials, and several drugs have been approved for the treatment of IBD. In the future, numerous potential emerging therapeutic options for IBD treatment are believed to come to the fore, achieving disease cure.
Collapse
Affiliation(s)
- Zhezhe Tian
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, China
- Hepatic Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qiaorui Zhao
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease−Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xiu Teng
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease−Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Liu J, Zhao J, Zhang YL, Zhang C, Yang GD, Tian WS, Zhou BH, Wang HW. Underlying Mechanism of Fluoride Inhibits Colonic Gland Cells Proliferation by Inducing an Inflammation Response. Biol Trace Elem Res 2024:10.1007/s12011-024-04212-6. [PMID: 38995434 DOI: 10.1007/s12011-024-04212-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 07/13/2024]
Abstract
The integrity of colonic gland cells is a prerequisite for normal colonic function and maintenance. To evaluate the underlying injury mechanisms in colonic gland cells induced by excessive fluoride (F), forty-eight female Kunming mice were randomly allocated into four groups and treated with different concentrations of NaF (0, 25, 50, and 100 mg F-/L) for 70 days. As a result, the integrity of the colonic mucosa and the cell layer was seriously damaged after F treatment, as manifested by atrophy of the colonic glands, colonic cell surface collapse, breakage of microvilli, and mitochondrial vacuolization. Alcian blue and periodic acid Schiff staining revealed that F decreased the number of goblet cells and glycoprotein secretion. Furthermore, F increased the protein expression of TLR4, NF-κB, and ERK1/2 and decreased IL-6, interfered with NF-κB signaling, following induced colonic gland cells inflammation. The accumulation of F inhibited proliferation via the JAK/STAT signaling pathway, as characterized by decreased mRNA and protein expression of JAK, STAT3, STAT5, PCNA, and Ki67 in colon tissue. Additionally, the expression of CDK4 was up-regulated by increased F concentration. In conclusion, excessive F triggered colonic inflammation and inhibited colonic gland cell proliferation via regulation of the NF-κB and JAK/STAT signaling pathways, leading to histopathology and barrier damage in the colon. The results explain the damaging effect of the F-induced inflammatory response on the colon from the perspective of cell proliferation and provide a new idea for explaining the potential mechanism of F-induced intestinal damage.
Collapse
Affiliation(s)
- Jing Liu
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Jing Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Yu-Ling Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Cai Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Guo-Dong Yang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Wei-Shun Tian
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Bian-Hua Zhou
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China.
| |
Collapse
|
45
|
Liang L, Zhang J, Chen J, Tian Y, Li W, Shi M, Cheng S, Zheng Y, Wang C, Liu H, Yang X, Ye W. Bazedoxifene attenuates dextran sodium sulfate-induced colitis in mice through gut microbiota modulation and inhibition of STAT3 and NF-κB pathways. Eur J Pharmacol 2024; 974:176611. [PMID: 38663540 DOI: 10.1016/j.ejphar.2024.176611] [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: 01/03/2024] [Revised: 03/26/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder of the gastrointestinal tract for which treatment options remain limited. In this study, we used a dual-luciferase-based screening of an FDA-approved drug library, identifying Bazedoxifene (BZA) as an inhibitor of the NF-κB pathway. We further investigated its therapeutic effects in a dextran sodium sulfate (DSS)-induced colitis model and explored its impact on gut microbiota regulation and the underlying molecular mechanisms. Our results showed that BZA significantly reduced DSS-induced colitis symptoms in mice, evidenced by decreased colon length shortening, lower histological scores, and increased expression of intestinal mucosal barrier-associated proteins, such as Claudin 1, Occludin, Zo-1, Mucin 2 (Muc2), and E-cadherin. Used independently, BZA showed therapeutic effects comparable to those of infliximab (IFX). In addition, BZA modulated the abundance of gut microbiota especially Bifidobacterium pseudolongum, and influenced microbial metabolite production. Crucially, BZA's alleviation of DSS-induced colitis in mice was linked to change in gut microbiota composition, as evidenced by in vivo gut microbiota depletion and fecal microbiota transplantation (FMT) mice model. Molecularly, BZA inhibited STAT3 and NF-κB activation in DSS-induced colitis in mice. In general, BZA significantly reduced DSS-induced colitis in mice through modulating the gut microbiota and inhibiting STAT3 and NF-κB activation, and its independent use demonstrated a therapeutic potential comparable to IFX. This study highlights gut microbiota's role in IBD drug development, offering insights for BZA's future development and its clinical applications.
Collapse
Affiliation(s)
- Liumei Liang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Jingdan Zhang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Junxiong Chen
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Yu Tian
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Weiqian Li
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Mengchen Shi
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Sijing Cheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China; Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China
| | - Yinhai Zheng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Chen Wang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Huanliang Liu
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China
| | - Xiangling Yang
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, 510655, China.
| | - Weibiao Ye
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510655, China; Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, 523059, China.
| |
Collapse
|
46
|
Zhang YF, Fan MY, Bai QR, Zhao R, Song S, Wu L, Lu JH, Liu JW, Wang Q, Li Y, Chen X. Precision therapy for ulcerative colitis: insights from mitochondrial dysfunction interacting with the immune microenvironment. Front Immunol 2024; 15:1396221. [PMID: 39026683 PMCID: PMC11254623 DOI: 10.3389/fimmu.2024.1396221] [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: 03/05/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Background Accumulating evidence reveals mitochondrial dysfunction exacerbates intestinal barrier dysfunction and inflammation. Despite the growing knowledge of mitochondrial dysfunction and ulcerative colitis (UC), the mechanism of mitochondrial dysfunction in UC remains to be fully explored. Methods We integrated 1137 UC colon mucosal samples from 12 multicenter cohorts worldwide to create a normalized compendium. Differentially expressed mitochondria-related genes (DE-MiRGs) in individuals with UC were identified using the "Limma" R package. Unsupervised consensus clustering was utilized to determine the intrinsic subtypes of UC driven by DE-MiRGs. Weighted gene co-expression network analysis was employed to investigate module genes related to UC. Four machine learning algorithms were utilized for screening DE-MiRGs in UC and construct MiRGs diagnostic models. The models were developed utilizing the over-sampled training cohort, followed by validation in both the internal test cohort and the external validation cohort. Immune cell infiltration was assessed using the Xcell and CIBERSORT algorithms, while potential biological mechanisms were explored through GSVA and GSEA algorithms. Hub genes were selected using the PPI network. Results The study identified 108 DE-MiRGs in the colonic mucosa of patients with UC compared to healthy controls, showing significant enrichment in pathways associated with mitochondrial metabolism and inflammation. The MiRGs diagnostic models for UC were constructed based on 17 signature genes identified through various machine learning algorithms, demonstrated excellent predictive capabilities. Utilizing the identified DE-MiRGs from the normalized compendium, 941 patients with UC were stratified into three subtypes characterized by distinct cellular and molecular profiles. Specifically, the metabolic subtype demonstrated enrichment in epithelial cells, the immune-inflamed subtype displayed high enrichment in antigen-presenting cells and pathways related to pro-inflammatory activation, and the transitional subtype exhibited moderate activation across all signaling pathways. Importantly, the immune-inflamed subtype exhibited a stronger correlation with superior response to four biologics: infliximab, ustekinumab, vedolizumab, and golimumab compared to the metabolic subtype. Conclusion This analysis unveils the interplay between mitochondrial dysfunction and the immune microenvironment in UC, thereby offering novel perspectives on the potential pathogenesis of UC and precision treatment of UC patients, and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Yi-fan Zhang
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Meng-ying Fan
- The Anesthesiology College, Shanxi Medical University, Taiyuan, China
| | - Qi-rui Bai
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shan Song
- The Second Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Li Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Jun-hui Lu
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing-wei Liu
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yuan Li
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xing Chen
- Department of Gastroenterology, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
47
|
Yokomizo T, Oshima M, Iwama A. Epigenetics of hematopoietic stem cell aging. Curr Opin Hematol 2024; 31:207-216. [PMID: 38640057 DOI: 10.1097/moh.0000000000000818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
PURPOSE OF REVIEW The development of new antiaging medicines is of great interest to the current elderly and aging population. Aging of the hematopoietic system is attributed to the aging of hematopoietic stem cells (HSCs), and epigenetic alterations are the key effectors driving HSC aging. Understanding the epigenetics of HSC aging holds promise of providing new insights for combating HSC aging and age-related hematological malignancies. RECENT FINDINGS Aging is characterized by the progressive loss of physiological integrity, leading to impaired function and increased vulnerability to death. During aging, the HSCs undergo both quantitative and qualitative changes. These functional changes in HSCs cause dysregulated hematopoiesis, resulting in anemia, immune dysfunction, and an increased risk of hematological malignancies. Various cell-intrinsic and cell-extrinsic effectors influencing HSC aging have also been identified. Epigenetic alterations are one such mechanism. SUMMARY Cumulative epigenetic alterations in aged HSCs affect their fate, leading to aberrant self-renewal, differentiation, and function of aged HSCs. In turn, these factors provide an opportunity for aged HSCs to expand by modulating their self-renewal and differentiation balance, thereby contributing to the development of hematological malignancies.
Collapse
Affiliation(s)
- Takako Yokomizo
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
48
|
Coronado-Rivera EF, Alcántara-Figueroa CE. Upadacitinib in a patient with Crohn's disease with loss of response to ustekinumab: A case report. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2024; 89:449-450. [PMID: 39034266 DOI: 10.1016/j.rgmxen.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/03/2024] [Indexed: 07/23/2024]
Affiliation(s)
- E F Coronado-Rivera
- Escuela de Medicina, Universidad Privada Antenor Orrego, Trujillo, Peru; Servicio de Anestesiología y Centro Quirúrgico, Hospital Víctor Lazarte Echegaray, Trujillo, Peru.
| | - C E Alcántara-Figueroa
- Escuela de Medicina, Universidad Privada Antenor Orrego, Trujillo, Peru; Servicio de Gastroenterología, Hospital Belén, Trujillo, Peru
| |
Collapse
|
49
|
Hammerhøj A, Chakravarti D, Sato T, Jensen KB, Nielsen OH. Organoids as regenerative medicine for inflammatory bowel disease. iScience 2024; 27:110118. [PMID: 38947526 PMCID: PMC11214415 DOI: 10.1016/j.isci.2024.110118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder with an increasing global prevalence. Managing disease activity relies on various pharmacological options. However, the effectiveness of current therapeutics is limited and not universally applicable to all patients and circumstances. Consequently, developing new management strategies is necessary. Recent advances in endoscopically obtained intestinal biopsy specimens have highlighted the potential of intestinal epithelial organoid transplantation as a novel therapeutic approach. Experimental studies using murine and human organoid transplantations have shown promising outcomes, including tissue regeneration and functional recovery. Human trials with organoid therapy have commenced; thus, this article provides readers with insights into the necessity and potential of intestinal organoid transplantation as a new regenerative therapeutic option in clinical settings and explores its associated challenges.
Collapse
Affiliation(s)
- Alexander Hammerhøj
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kim Bak Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
50
|
Watanabe K, Peyrin-Biroulet L, Danese S, Fujitani Y, Faes M, Oortwijn A, Lindsay JO, Rogler G, Hibi T. Impact of Concomitant Thiopurine on the Efficacy and Safety of Filgotinib in Patients with Ulcerative Colitis: Post Hoc Analysis of the Phase 2b/3 SELECTION Study. J Crohns Colitis 2024; 18:801-811. [PMID: 38019901 PMCID: PMC11147794 DOI: 10.1093/ecco-jcc/jjad201] [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: 06/25/2023] [Revised: 11/06/2023] [Accepted: 11/28/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND AND AIMS SELECTION is the first study to assess the impact of concomitant thiopurine and other immunomodulator [IM] use on the efficacy and safety of a Janus kinase inhibitor, filgotinib, in patients with ulcerative colitis. METHODS Data from the phase 2b/3 SELECTION study were used for this post hoc analysis. Patients were randomised [2:2:1] to two induction studies [biologic-naive, biologic-experienced] to filgotinib 200 mg, 100 mg, or placebo. At Week 10, patients receiving filgotinib were re-randomised [2:1] to continue filgotinib or to switch to placebo until Week 58 [maintenance]. Outcomes were compared between subgroups with and without concomitant IM use. RESULTS At Week 10, similar proportions of patients in the +IM and -IM groups treated with filgotinib 200 mg achieved Mayo Clinic Score [MCS] response [biologic-naive: 65.8% vs 66.9%; biologic-experienced: 61.3% vs 50.5%] and clinical remission [biologic-naive: 26.0% vs 26.2%; biologic-experienced: 11.3% vs 11.5%]. At Week 58, similar proportion of patients in the +IM and -IM groups treated with filgotinib 200 mg achieved MCS response [biologic-naive: 74.2% vs 75.0%; biologic-experienced: 45.5% vs 61.4%] and clinical remission [biologic-naive: 51.6% vs 47.4%; biologic-experienced: 22.7% vs 24.3%]. The probability of protocol-specified disease worsening during the maintenance study in patients treated with filgotinib 200 mg did not differ between +IM and -IM groups [p = 0.6700]. No differences were observed in the incidences of adverse events between +IM and -IM groups in the induction/maintenance studies. CONCLUSIONS The efficacy and safety profiles of filgotinib treatment in SELECTION did not differ with or without concomitant IM use. CLINICALTRIALS.GOV IDENTIFIER NCT02914522.
Collapse
Affiliation(s)
- Kenji Watanabe
- Department of Internal Medicine for Inflammatory Bowel Disease, University of Toyama, Toyama, Japan
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, CHRU-Nancy, University of Lorraine, Nancy, France
- Inserm, NGERE, University of Lorraine, Nancy, France
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Hospital San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | - James O Lindsay
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Toshifumi Hibi
- Center for Advanced IBD Research and Treatment, Kitasato Institute Hospital, Kitasato University, Tokyo, Japan
| |
Collapse
|