1
|
Chen KH, Xu R, Ye HJ, Xu B, Cao SL, Chen HX, Chen YL, Cai YP, Xie XQ, Zhao M, Zhou L, Luo X. Evaluating the efficacy and safety of emodin, luteolin, and paeonol combination from Dahuang Mudan decoction in ameliorating ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119692. [PMID: 40157404 DOI: 10.1016/j.jep.2025.119692] [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: 01/19/2025] [Revised: 03/11/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dahuang Mudan Decoction is a classic Chinese medicine prescription for treating ulcerative colitis (UC). Previous studies have shown that Dahuang Mudan Decoction has preventive and therapeutic effects on mice with dextran sulfate sodium (DSS) induced colitis. AIM OF THE STUDY The objective of this research endeavor was to ascertain the most efficacious synergistic blend of Emodin, Luteolin, and Paeonol, the main active ingredients in Dahuang Mudan Decoction, in alleviating UC. Additionally, it sought to elucidate the underlying therapeutic mechanisms and evaluate the safety of the combined components. MATERIALS AND METHODS Employing Emodin, Luteolin, and Paeonol as starting materials, the optimal combination was selected by orthogonal design. Basic pharmacodynamics was observed in mouse model of UC induced by DSS. The pathological changes of the colon were observed using hematoxylin and eosin (H&E) staining. The changes of cytokines and proteins related to inflammation and intestinal barrier function were detected by WB, Alcian blue staining, immunofluorescence, immunohistochemistry and related kits. Subsequently, 16S rRNA sequencing was used to observe changes in the intestinal flora. To evaluate the therapeutic effect and potential mechanism of the optimal monomer composition on UC mouse model. Finally, we performed toxicity tests as part of the safety assessment of the combination of the three monomers. RESULTS The different combinations of Emodin, Luteolin, and Paeonol alleviated DSS-induced colitis to varying degrees. The ELP5 group (Emodin 5 mg/kg + Luteolin 5 mg/kg + Paeonol 15 mg/kg) and ELP9 group (Emodin 15 mg/kg + Luteolin 15 mg/kg + Paeonol 75 mg/kg) had the most significant mitigation effect on UC mice. Mechanistically, the monomeric composition provides a comprehensive treatment for UC by addressing multiple aspects, including anti-inflammatory and antioxidant effects, repairing the damaged intestinal barrier, restoring the intestinal flora structure, and regulating short-chain fatty acid levels. In addition, the combination of Emodin, Luteolin and Paeonol exhibited a more significant effect on DSS-induced colitis compared to the individual components, indicating a synergistic effect among them. In the single-dose toxicity test, no obvious abnormalities were found in the general state or major organs of the mice. In repeated toxicity tests, it was found that the combined use of three monomers had less effect on organ index, hematology and serum biochemical indexes than that of a single compound. Pathological examination showed that the three monomers had certain toxicity to mouse liver, kidney and lung when used alone and in large doses for a long time, and the toxicity was significantly reduced after combined use. CONCLUSIONS We have determined the optimal combination of three active ingredients in Dahuang Mudan Decoction to alleviate DSS induced colitis in mice by inhibiting intestinal inflammation and oxidative stress, repairing impaired intestinal barrier function, and regulating intestinal flora disturbance. The results of single administration toxicity test proved the safety of the three monomers combined, and repeated administration toxicity test clarified the safe dose range of the combined administration, and also revealed that the combined therapy exhibited superior safety compared to monotherapy.
Collapse
MESH Headings
- Animals
- Emodin/pharmacology
- Emodin/administration & dosage
- Emodin/therapeutic use
- Emodin/toxicity
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/pathology
- Acetophenones/administration & dosage
- Acetophenones/pharmacology
- Acetophenones/toxicity
- Acetophenones/therapeutic use
- Luteolin/pharmacology
- Luteolin/administration & dosage
- Luteolin/therapeutic use
- Luteolin/toxicity
- Dextran Sulfate
- Mice
- Male
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/therapeutic use
- Drugs, Chinese Herbal/toxicity
- Disease Models, Animal
- Mice, Inbred C57BL
- Colon/drug effects
- Colon/pathology
- Drug Therapy, Combination
- Gastrointestinal Microbiome/drug effects
- Anti-Inflammatory Agents/pharmacology
Collapse
Affiliation(s)
- Ke-Han Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hua-Jian Ye
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shui-Ling Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hong-Xu Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun-Liang Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan-Ping Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue-Qian Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xia Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China; StateKey Laboratory of Traditional Chinese Medicine Syndrome, China.
| |
Collapse
|
2
|
Pan X, Xian P, Li Y, Zhao X, Zhang J, Song Y, Nan Y, Ni S, Hu K. Chemotaxis-driven hybrid liposomes recover intestinal homeostasis for targeted colitis therapy. J Control Release 2025; 380:829-845. [PMID: 39961435 DOI: 10.1016/j.jconrel.2025.02.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Inflammatory bowel disease (IBD) is closely linked to the dysregulation of intestinal homeostasis, accompanied by intestinal epithelial barrier destruction, dysbiosis of gut microbiota, subsequent inflammatory factor infiltration, and excessive oxidative stress. Conventional therapeutics focus on suppressing inflammation and often suffer from metabolic instability as well as limited targeting, thereby leading to suboptimal remission rates and severe side effects. Here, we designed bacterial outer membrane vesicle (OMV, from Stenotrophomonas maltophilia)-fused and borneol-modified liposomes (BO/OMV-lipo@LU) for targeted delivery of luteolin to recover intestinal homeostasis by alleviating inflammation and modulating dysregulated intestinal epithelial barrier, redox balance, and gut microbiota in IBD. In a Caco-2/HT29-MTX monolayer model, the OMV and borneol-bifunctionalized liposomes enhanced the uptake efficiency of unfunctionalized liposomes with a 2-fold increase. Owing to the chemotaxis-driven colon-targeting ability of OMVs and the ability of borneol to promote intestinal epithelial uptake, the hybrid liposomes successfully targeted the inflamed colon. In a colitis mouse model, BO/OMV-lipo@LU exhibited enhanced efficacy following oral administration. The BO/OMV-lipo@LU treatment increased the colon length and body weights of mice suffering colitis by 40 % and 15 %, respectively, with values comparable to the healthy control group. Notably, BO/OMV-lipo@LU alleviated proinflammatory markers, modulated redox balance, and restored the intestinal epithelial barrier. In addition, the formulation increased the abundance of beneficial microbiota while decreasing the abundance of harmful microbiota. These results demonstrated that this biomimetic nanoplatform could be exploited as a safe and effective gut-targeted delivery system in IBD treatment.
Collapse
Affiliation(s)
- Xier Pan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peng Xian
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yushu Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Zhao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaxin Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yangjie Song
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunrong Nan
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
Muro P, Jing C, Zhao Z, Jin T, Mao F. The emerging role of honeysuckle flower in inflammatory bowel disease. Front Nutr 2025; 12:1525675. [PMID: 40225345 PMCID: PMC11985448 DOI: 10.3389/fnut.2025.1525675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC), referred to as inflammatory bowel disease (IBD), pose considerable challenges in treatment because they are chronic conditions that easily relapse. The occurrence of IBD continues to rise in developing countries. Nonetheless, the existing therapies for IBD have limitations and fail to address the needs of the patients thoroughly. There is an increasing need for new, safe, and highly effective alternative medications for IBD patients. Traditional Chinese Medicine (TCM) is employed in drug development and disease management due to its wide-range of biological activities, minimal toxicity, and limited side effects. Extensive research has shown that certain TCM exhibits significant therapeutic benefits for IBD treatments. Honeysuckle (Lonicera japonica) was used in TCM research and clinical settings for the treatment of IBD. Bioactive metabolites in L. japonica, such as luteolin, quercetin, cyanidin, chlorogenic acid (CGA), caffeic acid (CA), and saponin, exhibit significant therapeutic benefits for managing IBD. The honeysuckle flower is a potential candidate in the treatment of IBD due to its anti-inflammatory, immune system-regulating, and antioxidant qualities. This paper reviews the metabolites of the honeysuckle flower as a candidate for the treatment of IBD. It discusses the fundamental mechanism of L. japonica and the potential of its bioactive metabolites in the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Peter Muro
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Caihong Jing
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Zhihan Zhao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
4
|
Su Y, Feng C, Ye W, Xiao J, Meng Q, Yang X, Wang Y, Huang T, Lan L, Chen S, Ding Z, Su S, Wei S, Shan Q. Exploring the dynamic responses of group 3 innate lymphoid cells at different times in response to LPS challenge. Int Immunopharmacol 2025; 148:114162. [PMID: 39889415 DOI: 10.1016/j.intimp.2025.114162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Group 3 innate lymphoid cells (ILC3s) have clear roles in regulating mucosal immunity and tissue homeostasis in the intestine, though the immunological functions in lungs remain unclear. This study aimed to demonstrate the dynamic responses of ILC3s to acute inflammation upon LPS challenge. Microarray data and single-cell RNA sequencing (scRNA-seq) data obtained from the GEO database were combined to analyze the function of ILC3 subset, confirmed by flow cytometry assay and qRT-PCR. The gene enrichment analysis of intersected genes identified between microarray data in bacterial pneumonia and single-cell RNA sequencing of intestinal ILC3s were closely related to TNF-alpha effects on cytokine activity, cell motility and apoptosis pathway, indicating the possibility of intestinal ILC3s migration to the lung. Furthermore, the cellular landscapes of ILC3s in lung and intestine at different times after pulmonary infection exhibited varied ILC3 statuses. ILC3s in lung expanded a lot at 48 h while intestinal ILC3s decreased at 72 h response to LPS challenge, with higher expression of marked genes related to TNF-alpha effects on cytokine activity, cell motility and apoptosis pathway. The main findings in our study may serve as valuable resources for understanding the roles that ILC3s play upon LPS challenge, which may offer opportunities for translating ILC3s as therapeutic targets to regulate LPS-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Ying Su
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Caixia Feng
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Wenyu Ye
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Juan Xiao
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Qi Meng
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Xia Yang
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Yongcai Wang
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Ting Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture Guangxi Academy of Fishery Sciences Nanning China
| | - Liancheng Lan
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Sixing Chen
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Ziting Ding
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Shiqi Su
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Sumei Wei
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Qingwen Shan
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China.
| |
Collapse
|
5
|
Qu Q, Ma YM, Zhang WB, Chen R, Wang ZH, Jin WX, Huang YW, Xuan ZY, Liu MJ, Chen XL, Lv WJ, Guo SN. Period circadian clock 3 is crucial for regulation of IL-22-producing type 3 innate lymphoid cells by flavonoids from Shen Ling Bai Zhu San to alleviate colitis. Int J Biol Macromol 2025; 288:138730. [PMID: 39672410 DOI: 10.1016/j.ijbiomac.2024.138730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/15/2024]
Abstract
Type 3 Innate lymphoid cells (ILC3s) functions bear complex response during Inflammatory bowel diseases (IBD). Here, our study first analyzed the main pharmacological components in Shen Ling Bai Zhu San n-butanol extracts (S-Nb), and then explored whether S-Nb administrated immune response of ILC3s, and how it regulates ILC3s. Shen Ling Bai Zhu San (SLBZS) or S-Nb were administrated for 7 days to analyze the frequency of ILC3s and their produced cytokine. Using siRNA technology to knock down the expression of period circadian clock 2 (Per2) and period circadian clock 3 (Per3) and Anti-IL-22 antibody was supplied to mice, then detecting the moderator effect of S-Nb on colitis. The most class of S-Nb is flavonoids, with a content of approximately 48%. Oral administration of S-Nb enhanced the production of NCR+ILC3s and IL-22 produced by ILC3s, but did not alter IL-17A. Surprisingly, knocking down the expression of Per3 instead of Per2 inhibited the modulation effect of S-Nb on colitis and reduced IL-22 production, whether originating from NCR+ILC3s or NCR-ILC3s. After neutralizing the expression of IL-22 in mice, S-Nb was deprived of ability to alleviate colitis. The reason why S-Nb alleviates colitis is by enhancing the expression of Per3 via flavonoids, which in turn promotes the secretion of IL-22+ILC3s in intestine.
Collapse
Affiliation(s)
- Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yi-Mu Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Wen-Bo Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Rong Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhi-Hua Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Wen-Xin Jin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yi-Wen Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhao-Ying Xuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Meng-Jie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiao-Li Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Wei-Jie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| | - Shi-Ning Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong 510642, China; Guangdong Research Center for Veterinary Traditional Chinese Medicine and Natural Medicine Engineering Technology, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
6
|
Yang S, Duan H, Zeng J, Yan Z, Niu T, Ma X, Zhang Y, Hu J, Zhang L, Zhao X. Luteolin modulates macrophage phenotypic switching via the AMPK-PPARγ pathway to alleviate ulcerative colitis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 339:119157. [PMID: 39603400 DOI: 10.1016/j.jep.2024.119157] [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: 09/14/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lonicerae japonicae flos (LJF), the dried flower bud or newly bloomed flower of Lonicera japonica Thunb., is widely used in Traditional Chinese medicine (TCM), exhibiting anti-inflammatory and immune-enhancing properties. Luteolin (Lut) is a major bioactive component of LJF, demonstrating a regulatory role in immune disorders. However, the specific role of Lut in regulating macrophage-mediated intestinal inflammation and its underlying molecular mechanisms have not yet been fully explored. AIM OF THE STUDY This study was designed to explore whether Lut alleviates Ulcerative colitis (UC) in mice and to elucidate its underlying mechanism in intestinal inflammation. MATERIALS AND METHODS Mice were administered Dextran sodium sulfate (DSS) for 7 d to establish a UC model, followed by oral administration of Lut (12.5, 25, and 50 mg/kg body weight). RNA-sequencing (RNA-Seq) was used to screen signaling pathways. RAW264.7 cells were cultured and treated with Lut (6.25, 12.5, and 25 μM) and lipopolysaccharide (LPS, 1 μg/mL) for 24 h. To examine the role of the AMP-activated protein kinase (AMPK)/Peroxisome proliferator-activated receptor γ (PPARγ) signaling pathway, the cells were treated with compound C (an AMPK inhibitor) and GW9662 (a PPARγ antagonist). RESULTS Lut suppressed the inflammation of DSS-induced colitis in vivo, attenuated DSS-induced clinical man-ifestations, reversed colon length reduction, and reduced histological injury. Lut induced a shift in the macrophage phenotype from classical (M1) to alternative (M2) by suppressing M1 marker gene expression and enhancing M2 marker gene expression following DSS or LPS induction. RNA-seq revealed that PPARγ was involved in the regulation of macrophages by Lut. Furthermore, the polarization effect of Lut on macrophages was shown to be mediated through the AMPK-PPARγ signaling pathway. CONCLUSION These findings indicate that Lut effectively ameliorates UC in mice through the activation of the AMPK-PPARγ signaling pathway, leading to the inhibition of macrophage M1 polarization and promotion of M2 polarization. This study provides insight into future research on the utilization of Lut-rich TCM dietary supplements as a prophylactic treatment strategy in the prevention of UC.
Collapse
Affiliation(s)
- Shuai Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Hongwei Duan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Jianlin Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Zhenxing Yan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Tian Niu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Xiaofei Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Junjie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China
| | - Lihong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China.
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730070, Gansu, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou, 730070, Gansu, China.
| |
Collapse
|
7
|
Berkinbayeva M, Gu W, Chen Z, Gao P. Group 3 Innate Lymphoid Cells: A Potential Therapeutic Target for Steroid Resistant Asthma. Clin Rev Allergy Immunol 2024; 68:1. [PMID: 39751959 PMCID: PMC11698894 DOI: 10.1007/s12016-024-09012-3] [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] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Asthma is a chronic airway inflammatory disease that affects millions globally. Although glucocorticoids are a mainstay of asthma treatment, a subset of patients show resistance to these therapies, resulting in poor disease control and increased morbidity. The complex mechanisms underlying steroid-resistant asthma (SRA) involve Th1 and Th17 lymphocyte activity, neutrophil recruitment, and NLRP3 inflammasome activation. Recent studies provided evidence that innate lymphoid cells type 3 (ILC3s) might be potential therapeutic targets for non-eosinophilic asthma (NEA) and SRA. Like Th17 cells, ILC3s play crucial roles in immune responses, inflammation, and tissue homeostasis, contributing to disease severity and corticosteroid resistance in NEA. Biologics targeting ILC3-related pathways have shown promise in managing Th2-low asthma, suggesting new avenues for SRA treatment. This review aims to explore the risk factors for SRA, discuss the challenges and mechanisms underlying SRA, consolidate current findings on innate lymphoid cells, and elucidate their role in respiratory conditions. We present the latest findings on the involvement of ILC3s in human diseases and explore their potential mechanisms in SRA development. Furthermore, we review emerging therapeutic biologics targeting ILC3-related pathways in managing NEA and SRA. This review highlights current challenges, and emerging therapeutic strategies, and addresses a significant gap in asthma research, with implications for improving the management of steroid-resistant asthma.
Collapse
Affiliation(s)
- Marzhan Berkinbayeva
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
| | - Wenjing Gu
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhifeng Chen
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA.
| |
Collapse
|
8
|
Gao X, Feng X, Hou T, Huang W, Ma Z, Zhang D. The roles of flavonoids in the treatment of inflammatory bowel disease and extraintestinal manifestations: A review. FOOD BIOSCI 2024; 62:105431. [DOI: 10.1016/j.fbio.2024.105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Xie X, Li P, Zhao M, Xu B, Zhang G, Wang Q, Ni C, Luo X, Zhou L. Luteolin ameliorates ulcerative colitis in mice via reducing the depletion of NCR +ILC3 through Notch signaling pathway. Chin J Nat Med 2024; 22:991-1002. [PMID: 39510641 DOI: 10.1016/s1875-5364(24)60568-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Indexed: 11/15/2024]
Abstract
The disorder of group 3 innate lymphoid cells (ILC3) subgroup, such as the predominance of NCR-ILC3 but the depletion of NCR+ILC3, is unfavorable to damaged intestinal barrier repair, which leads to the prolongations and obstinacy of ulcerative colitis (UC). Our previous studies had shown that luteolin promoted NCR-ILC3 differentitating into NCR+ILC3 to improving the depletion of NCR+ILC3 in UC mice, while the mechanism is unclear. This article aimed to explore the underlying mechanism of luteolin enhancing the proportion NCR+ILC3. UC mice model was established with 2% DSS and Notch signaling was blocked, then luteolin was used to intervene. The results showed that the effect of luteolin on ameliorating disease symptoms in UC mice, including inhibiting the weight loss, reducing the pathological damage of colon mucosa, etc., was diminished with blocking Notch signaling pathway. In addition, luteolin increased the proportion of NCR+ILC3, NCR+MNK3 and IL-22+ILC3, decreased intestinal permeability, promoted mucin secretion, and promoted ZO-1 and Occludin expression, the above effect of luteolin was neutralized by Notch inhibitor LY-411575. Luteolin activated the abnormally blocked Notch signaling pathway in UC mice. And molecular docking predicted the affinity of luteolin for RBPJ to be -7.5 kcal·mol-1 in mouse, respectively; the affinity of luteolin for Notch1 and RBPJ was respectively scored to be -6.4 kcal·mol-1 and -7.7 kcal·mol-1 homo sapiens. These results proved that luteolin is positive for enhancing the proportion of NCR+ILC3 via Notch signaling, and it provides a basis for targeting NCR+ILC3 for restoring intestinal barrier function to alleviating ulcerative colitis.
Collapse
Affiliation(s)
- Xueqian Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Pengcheng Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Meng Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Bo Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Guixing Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen 518000, China
| | - Qing Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Chen Ni
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China
| | - Xia Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| | - Lian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510000, China.
| |
Collapse
|
10
|
Huang Y, Wu Q, Li S, Lin X, Yang S, Zhu R, Fu C, Zhang Z. Harnessing nature's pharmacy: investigating natural compounds as novel therapeutics for ulcerative colitis. Front Pharmacol 2024; 15:1394124. [PMID: 39206263 PMCID: PMC11349575 DOI: 10.3389/fphar.2024.1394124] [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: 03/06/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024] Open
Abstract
Backgrounds Ulcerative colitis (UC) is a form of chronic inflammatory bowel disease, and UC diagnosis rates continue to rise throughout the globe. The research and development of new drugs for the treatment of UC are urgent, and natural compounds are an important source. However, there is a lack of systematic summarization of natural compounds and their mechanisms for the treatment of UC. Methods We reviewed the literature in the databases below from their inception until July 2023: Web of Science, PubMed, China National Knowledge Infrastructure, and Wanfang Data, to obtain information on the relationship between natural compounds and UC. Results The results showed that 279 natural compounds treat UC through four main mechanisms, including regulating gut microbiota and metabolites (Mechanism I), protecting the intestinal mucosal barrier (Mechanism II), regulating intestinal mucosal immune response (Mechanism III), as well as regulating other mechanisms (Mechanism Ⅳ) such as cellular autophagy modulation and ferroptosis inhibition. Of these, Mechanism III is regulated by all natural compounds. The 279 natural compounds, including 62 terpenoids, 57 alkaloids, 52 flavonoids, 26 phenols, 19 phenylpropanoids, 9 steroids, 9 saponins, 8 quinonoids, 6 vitamins, and 31 others, can effectively ameliorate UC. Of these, terpenoids, alkaloids, and flavonoids have the greatest potential for treating UC. It is noteworthy to highlight that a total of 54 natural compounds exhibit their therapeutic effects by modulating Mechanisms I, II, and III. Conclusion This review serves as a comprehensive resource for the pharmaceutical industry, researchers, and clinicians seeking novel therapeutic approaches to combat UC. Harnessing the therapeutic potential of these natural compounds may significantly contribute to the improvement of the quality of life of patients with UC and promotion of disease-modifying therapies in the future.
Collapse
Affiliation(s)
- You Huang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuhong Wu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sha Li
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xia Lin
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shasha Yang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Zhu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomei Fu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Zhang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Hanlon N, Gillan N, Neil J, Seidler K. The role of the aryl hydrocarbon receptor (AhR) in modulating intestinal ILC3s to optimise gut pathogen resistance in lupus and benefits of nutritional AhR ligands. Clin Nutr 2024; 43:1199-1215. [PMID: 38631087 DOI: 10.1016/j.clnu.2024.04.008] [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: 09/18/2023] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND AND AIMS Dysbiosis is emerging as a potential trigger of systemic lupus erythematosus (SLE). Group 3 innate lymphoid cells (ILC3s) are recognised as key regulators of intestinal homeostasis. The aryl hydrocarbon receptor (AhR) is critical to intestinal ILC3 development and function. This mechanistic review aimed to investigate whether AhR activation of gut ILC3s facilitates IL-22-mediated antimicrobial peptide (AMP) production to enhance colonisation resistance and ameliorate SLE pathology associated with intestinal dysbiosis. Furthermore, nutritional AhR ligand potential to enhance pathogen resistance was explored. METHODOLOGY This mechanistic review involved a three-tranche systematic literature search (review, mechanism, intervention) using PubMed with critical appraisal. Data was synthesised into themes and summarised in a narrative analysis. RESULTS Preclinical mechanistic data indicate that AhR modulation of intestinal ILC3s optimises pathogen resistance via IL-22-derived AMPs. Pre-clinical research is required to validate this mechanism in SLE. Data on systemic immune consequences of AhR modulation in lupus suggest UVB-activated ligands induce aberrant AhR signalling while many dietary ligands exert beneficial effects. Data on xenobiotic-origin ligands is varied, although considerable evidence has demonstrated negative effects on Th17 to Treg balance. Limited human evidence supports the role of nutritional AhR ligands in modulating SLE pathology. Preclinical and clinical data support anti-inflammatory effects of dietary AhR ligands. CONCLUSION Current evidence is insufficient to fully validate the hypothesis that AhR modulation of intestinal ILC3s can enhance pathogen resistance to ameliorate lupus pathology driven by dysbiosis. However, anti-inflammatory effects of dietary AhR ligands suggest a promising role as a therapeutic intervention for SLE.
Collapse
Affiliation(s)
- Niamh Hanlon
- CNELM (Centre for Nutrition Education and Lifestyle Management), 14 Rectory Road, Wokingham, Berkshire RG40 1DH, UK.
| | - Natalie Gillan
- CNELM (Centre for Nutrition Education and Lifestyle Management), 14 Rectory Road, Wokingham, Berkshire RG40 1DH, UK.
| | - James Neil
- CNELM (Centre for Nutrition Education and Lifestyle Management), 14 Rectory Road, Wokingham, Berkshire RG40 1DH, UK.
| | - Karin Seidler
- CNELM (Centre for Nutrition Education and Lifestyle Management), 14 Rectory Road, Wokingham, Berkshire RG40 1DH, UK.
| |
Collapse
|
12
|
Bai J, Wang Y, Li F, Wu Y, Chen J, Li M, Wang X, Lv B. Research advancements and perspectives of inflammatory bowel disease: A comprehensive review. Sci Prog 2024; 107:368504241253709. [PMID: 38778725 PMCID: PMC11113063 DOI: 10.1177/00368504241253709] [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] [Indexed: 05/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease with increasing incidence, such as Crohn's disease and ulcerative colitis. The accurate etiology and pathogenesis of IBD remain unclear, and it is generally believed that it is related to genetic susceptibility, gut microbiota, environmental factors, immunological abnormalities, and potentially other factors. Currently, the mainstream therapeutic drugs are amino salicylic acid agents, corticosteroids, immunomodulators, and biological agents, but the remission rates do not surpass 30-60% of patients in a real-life setting. As a consequence, there are many studies focusing on emerging drugs and bioactive ingredients that have higher efficacy and long-term safety for achieving complete deep healing. This article begins with a review of the latest, systematic, and credible summaries of the pathogenesis of IBD. In addition, we provide a summary of the current treatments and drugs for IBD. Finally, we focus on the therapeutic effects of emerging drugs such as microRNAs and lncRNAs, nanoparticles-mediated drugs and natural products on IBD and their mechanisms of action.
Collapse
Affiliation(s)
- Junyi Bai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Ying Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Fuhao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueyao Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Meng Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
13
|
Liang X, Zheng S, Zhou Y, Li J, Zhang Z. Luteolin, a natural flavonoid, exhibits a protective effect on intestinal injury induced by soybean meal in early-weaned piglets. J Anim Sci 2024; 102:skae214. [PMID: 39066584 PMCID: PMC11367559 DOI: 10.1093/jas/skae214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024] Open
Abstract
Soybean meal is known to be able to cause intestinal damage and dysfunction in early-weaned piglets. However, research on natural compounds that can alleviate these effects is scarce. In this study, the effect of luteolin, a natural flavonoid, on intestinal health of piglets fed on a soybean meal-based diet was explored. A total of eighteen 21-d-old piglets were selected and randomly divided into 3 groups: a negative control group fed with an animal protein-based diet, a positive control group fed with a soybean meal-based diet, and a luteolin group that was fed with the positive control diet supplemented with luteolin. The results suggested that luteolin supplementation significantly increased the average daily gain and average daily feed intake of early-weaned piglets, while effectively reducing the diarrhea incidence. Additionally, luteolin supplementation lowered the levels of soybean antigen-specific immunoglobulin G and immunoglobulin E anitbodies, increased the superoxide dismutase activity in both sera and small intestine mucosa, and enhanced the total antioxidant capacity in sera. Further research found that luteolin supplementation increased the intestinal villi height and decreased the crypt depth, resulting in an increased ratio of villi to crypts. At the same time, it reduced the concentration of serum diamine oxidase, improving intestinal barrier function. Moreover, luteolin significantly decreased the gene expression of Bax and Caspase-3, reducing cell apoptosis in the intestinal mucosa. Luteolin supplementation also increased the abundance of Actinobacteria at the phylum level, reduced the abundance of Prevotella and increased the abundance of Olsenella at the genus level. In conclusion, the supplementation of luteolin to the soybean meal diet was capable of effectively reducing allergic response, enhancing the antioxidant capacity of early-weaned piglets, protecting their intestinal barrier function, inhibiting intestinal mucosal cell apoptosis, and altering the intestinal microbiota structure, therefore promoting intestinal health and improving production performance in early-weaned piglets.
Collapse
Affiliation(s)
- Xuecong Liang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning 110866, PR China
| | - Shugui Zheng
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning 110866, PR China
| | - Yang Zhou
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning 110866, PR China
| | - Jiguang Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning 110866, PR China
| | - Zhuo Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning 110866, PR China
| |
Collapse
|
14
|
Afsar S, Syed RU, Bin Break MK, Alsukaybi RH, Alanzi RA, Alshobrmi AM, Alshagdali NM, Alshammari AD, Alharbi FM, Alshammari AM, Algharbi WF, Albrykan KM, Alshammari FN. The dual role of MiR-210 in the aetiology of cancer: A focus on hypoxia-inducible factor signalling. Pathol Res Pract 2024; 253:155018. [PMID: 38070222 DOI: 10.1016/j.prp.2023.155018] [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: 10/19/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024]
Abstract
Tumorigenesis exemplifies the complex process of neoplasm origination, which is characterised by somatic genetic alterations and abnormal cellular growth. This multidimensional phenomenon transforms previously dormant cells into malignant equivalents, resulting in uncontrollable proliferation and clonal expansion. Various elements, including random mutations, harmful environmental substances, and genetic predispositions, influence tumorigenesis's aetiology. MicroRNAs (miRNAs) are now recognised as crucial determinants of gene expression and key players in several biological methods, including oncogenesis. A well-known hypoxia-inducible miRNA is MiR-210, which is of particular interest because of its complicated role in the aetiology of cancer and a variation of physiological and pathological situations. MiR-210 significantly impacts cancer by controlling the hypoxia-inducible factor (HIF) signalling pathway. By supporting angiogenesis, metabolic reprogramming, and cellular survival in hypoxic microenvironments, HIF signalling orchestrates adaptive responses, accelerating the unstoppable development of tumorous growth. Targeting several components of this cascade, including HIF-1, HIF-3, and FIH-1, MiR-210 plays a vital role in modifying HIF signalling and carefully controlling the HIF-mediated response and cellular fates in hypoxic environments. To understand the complexities of this relationship, careful investigation is required at the intersection of MiR-210 and HIF signalling. Understanding this relationship is crucial for uncovering the mechanisms underlying cancer aetiology and developing cutting-edge therapeutic approaches. The current review emphasises MiR-210's significance as a vital regulator of the HIF signalling cascade, with substantial implications spanning a range of tumor pathogenesis.
Collapse
Affiliation(s)
- S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh 517502, India
| | - Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia.
| | - Mohammed Khaled Bin Break
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia; Medical and Diagnostic Research Centre, University of Hail, Hail 55473, Saudi Arabia
| | | | - Reem A Alanzi
- College of Pharmacy, University of Hail, Hail 81442, Saudi Arabia
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Koprivica I, Stanisavljević S, Mićanović D, Jevtić B, Stojanović I, Miljković Đ. ILC3: a case of conflicted identity. Front Immunol 2023; 14:1271699. [PMID: 37915588 PMCID: PMC10616800 DOI: 10.3389/fimmu.2023.1271699] [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: 08/02/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
Innate lymphoid cells type 3 (ILC3s) are the first line sentinels at the mucous tissues, where they contribute to the homeostatic immune response in a major way. Also, they have been increasingly appreciated as important modulators of chronic inflammatory and autoimmune responses, both locally and systemically. The proper identification of ILC3 is of utmost importance for meaningful studies on their role in immunity. Flow cytometry is the method of choice for the detection and characterization of ILC3. However, the analysis of ILC3-related papers shows inconsistency in ILC3 phenotypic definition, as different inclusion and exclusion markers are used for their identification. Here, we present these discrepancies in the phenotypic characterization of human and mouse ILC3s. We discuss the pros and cons of using various markers for ILC3 identification. Furthermore, we consider the possibilities for the efficient isolation and propagation of ILC3 from different organs and tissues for in-vitro and in-vivo studies. This paper calls upon uniformity in ILC3 definition, isolation, and propagation for the increased possibility of confluent interpretation of ILC3's role in immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Jing S, Chen H, Liu E, Zhang M, Zeng F, Shen H, Fang Y, Muhitdinov B, Huang Y. Oral pectin/oligochitosan microspheres for colon-specific controlled release of quercetin to treat inflammatory bowel disease. Carbohydr Polym 2023; 316:121025. [PMID: 37321723 DOI: 10.1016/j.carbpol.2023.121025] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/29/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic, life quality-reducing disease with no cures available yet. To develop an effective medication suitable for long-term use is an urgent but unmet need. Quercetin (QT) is a natural dietary flavonoid with good safety and multifaceted pharmacological activities against inflammation. However, orally administrated quercetin yields unproductive outcomes for IBD treatment because of its poor solubility and extensive metabolism in the gastrointestinal tract. In this work, a colon-targeted QT delivery system (termed COS-CaP-QT) was developed, of which the pectin (PEC)/Ca2+ microspheres were prepared and then crosslinked by oligochitosan (COS). The drug release profile of COS-CaP-QT was pH-dependent and colon microenvironment-responsive, and COS-CaP-QT showed preferential distribution in the colon. The mechanism study showed that QT triggered the Notch pathway to regulate the proliferation of T helper 2 (Th2) cells and group 3 innate lymphoid cells (ILC3s) and the inflammatory microenvironment was remodeled. The in vivo therapeutic results revealed that COS-CaP-QT could relieve the colitis symptoms and maintain the colon length and intestinal barrier integrity.
Collapse
Affiliation(s)
- Shisuo Jing
- School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Huayuan Chen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Feng Zeng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510450, China
| | - Huan Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Shanghai Institute of Materia Medica, CAS, Shanghai 201203, China
| | - Yuefei Fang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Bahtiyor Muhitdinov
- Shanghai Institute of Materia Medica, CAS, Shanghai 201203, China; Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, Tashkent 100125, Uzbekistan
| | - Yongzhuo Huang
- School of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China; Shanghai Institute of Materia Medica, CAS, Shanghai 201203, China.
| |
Collapse
|
17
|
Wu P, Chang C, Zhu G, Zhai L, Zhang X, Huan Q, Gao Z, Deng H, Liang Y, Xiao H. Network Pharmacology Study of Bioactive Components and Molecular Mechanisms of the Glycoside Fraction from Picrorhiza scrophulariiflora Against Experimental Colitis. Drug Des Devel Ther 2023; 17:1531-1546. [PMID: 37249930 PMCID: PMC10224697 DOI: 10.2147/dddt.s407339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/29/2023] [Indexed: 05/31/2023] Open
Abstract
Purpose To explore the potential mechanism of glycosidic fraction of Picrorhiza scrophulariiflora Pennell (GPS) extract for the treatment of colitis using UPLC-QTOF-MS analysis, network pharmacology and experimental research. Methods The active components of GPS extract were identified by UPLC-QTOF-MS analysis and extracted their targets from the databases, which was used for network pharmacology analysis. Kyoto Encyclopedia of genes and genomes (KEGG) pathway analysis was performed to discover potential therapeutic mechanisms, and the network pharmacology results were then validated by in vivo and in vitro experiments. Results The results showed that GPS extract significantly alleviated the clinical signs of colitis, including body weight, disease activity index, colon shortening, and colon tissue damage, and inhibited the transcription and production of colonic IL-1β and IL-6 in DSS-induced colitis mice. In vitro, GPS extract also significantly suppressed nitric oxide (NO) production, iNOS expression, IL-1β and IL-6 transcription of LPS-activated RAW 264.7 cells. Network pharmacology integrated with experimental validation identified that GPS extract significantly suppressed Akt, p38, ERK, and JNK phosphorylation in vivo and in vitro, and luteolin, apocynin, caffeic acid, caffeic acid methyl ester, luteoloside, picroside II, aucubin, cinnamic acid, vanillic acid, and sweroside were the main components responsible for the anti-inflammatory effect of GPS. These findings demonstrate that the potential anti-inflammatory effect of GPS extract against colitis is achieved through suppressing PI3K/Akt and MAPK pathways, and that the abovementioned active components mainly exerted its anti-inflammatory effect. Conclusion The therapeutic effect of GPS extract on colitis is related to PI3K/Akt and MAPK pathways, which is a promising remedy for colitis therapy.
Collapse
Affiliation(s)
- Peigen Wu
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guizhou, People’s Republic of China
| | - Churui Chang
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guizhou, People’s Republic of China
| | - Guanglin Zhu
- Traditional Chinese Medicine Hospital of Qijiang, Chongqing, People’s Republic of China
| | - Lixiang Zhai
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong Special Administrative Region, People’s Republic of China
| | - Xu Zhang
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guizhou, People’s Republic of China
| | - Qiuchan Huan
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guizhou, People’s Republic of China
| | - Zhengxian Gao
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guizhou, People’s Republic of China
| | - Huan Deng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, People’s Republic of China
| | - Yue Liang
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, People’s Republic of China
| | - Haitao Xiao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, People’s Republic of China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guizhou, People’s Republic of China
| |
Collapse
|