1
|
Wu D, Huang Q, Xu Y, Cao R, Yang M, Xie J, Zhang D. Clinical efficacy and future application of indigo naturalis in the treatment of ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118782. [PMID: 39236777 DOI: 10.1016/j.jep.2024.118782] [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: 04/01/2024] [Revised: 08/03/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by non-specific inflammation. Managing UC presents significant challenges due to its chronic nature and high recurrence rates. Indigo naturalis has emerged as a potential therapeutic agent in clinical UC treatment, demonstrating advantages in alleviating refractory UC and maintaining remission periods compared to other therapeutic approaches. AIM OF REVIEW This review aims to elucidate the potential mechanisms underlying the therapeutic effects of indigo naturalis in UC treatment, assess its clinical efficacy, advantages, and limitations, and provide insights into methods and strategies for utilizing indigo naturalis in UC management. MATERIALS AND METHODS Comprehensive data on indigo naturalis were collected from reputable online databases including PubMed, GreenMedical, Web of Science, Google Scholar, China National Knowledge Infrastructure Database, and National Intellectual Property Administration. RESULTS Clinical studies have demonstrated that indigo naturalis, either alone or in combination with other drugs, yields favorable outcomes in UC treatment. Its mechanisms of action involve modulation of the AHR receptor, anti-inflammatory properties, regulation of intestinal flora, restoration of the intestinal barrier, and modulation of immunity. Despite its efficacy in managing refractory UC and prolonging remission periods, indigo naturalis treatment is associated with adverse reactions, quality variations, and inadequate pharmacokinetic investigations. CONCLUSION The therapeutic effects of indigo naturalis in UC treatment are closely linked to its ability to regulate the AHR receptor, exert anti-inflammatory effects, mcodulate intestinal flora, restore the intestinal barrier, and regulate immunity. Addressing the current shortcomings, including adverse reactions, quality control issues, and insufficient pharmacokinetic data, is crucial for optimizing the clinical utility of indigo naturalis in UC management. By refining patient-centered treatment strategies, indigo naturalis holds promise for broader application in UC treatment, thereby alleviating the suffering of UC patients.
Collapse
Affiliation(s)
- Dianzhen Wu
- Sichuan Medical Products Administration, Chengdu, 610017, China
| | - Qi Huang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yingbi Xu
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruiyi Cao
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ming Yang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Jin Xie
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Dingkun Zhang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Sichuan Provincial Engineering Research Center of Innovative Re-development of Famous Classical Formulas, Tianfu TCM Innovation Harbour, Chengdu University of Traditional Chinese Medicine, Chengdu, 611930, China.
| |
Collapse
|
2
|
Huang FC. Therapeutic Potential of Nutritional Aryl Hydrocarbon Receptor Ligands in Gut-Related Inflammation and Diseases. Biomedicines 2024; 12:2912. [PMID: 39767818 PMCID: PMC11673835 DOI: 10.3390/biomedicines12122912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
A solid scientific foundation is required to build the concept of personalized nutrition developed to promote health and a vision of disease prevention. Growing evidence indicates that nutrition can modulate the immune system through metabolites, which are either generated via microbiota metabolism or host digestion. The aryl hydrocarbon receptor (AhR) plays a crucial role in regulating immune responses, particularly in the gut, and has emerged as a key modulator of gut-mediated inflammation and related diseases. AhR is a ligand-activated transcription factor that responds to environmental, dietary, and microbial-derived signals, influencing immune balance and maintaining intestinal homeostasis. Nutritional AhR ligands play a significant role in modulating intestinal immunity and the function of mucosal immune cells, thereby exerting clinical effects on colitis and innate immunity. Additionally, they have the capacity to orchestrate autophagy, phagocytic cell function, and intestinal epithelial tight junctions. Therapeutic strategies aimed at enhancing AhR activity, restoring gut integrity, and optimizing immune responses hold promise as avenues for future research and potential treatments for critically ill patients.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| |
Collapse
|
3
|
Hu Y, Chen LL, Ye Z, Li LZ, Qian HZ, Wu MQ, Wang J, Qin KH, Ye QB. Indigo naturalis as a potential drug in the treatment of ulcerative colitis: a comprehensive review of current evidence. PHARMACEUTICAL BIOLOGY 2024; 62:818-832. [PMID: 39475104 PMCID: PMC11533244 DOI: 10.1080/13880209.2024.2415652] [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: 04/25/2024] [Revised: 08/25/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024]
Abstract
CONTEXT Ulcerative colitis (UC) is an intractable inflammatory bowel disease that threatens the health of patients. The limited availability of therapeutic strategies makes it imperative to explore more efficient and safer drugs. Indigo naturalis (IN) is a traditional Chinese medicine that possesses many pharmacological activities, including anti-inflammatory, antioxidant, and immunomodulatory activities. The treatment potential of IN for UC has been proven by numerous preclinical and clinical studies in recent years. OBJECTIVE This article provides a comprehensive review of the utility and potential of IN in the treatment of UC. METHODS 'Indigo naturalis' 'Qing dai' 'Qingdai' 'Ulcerative colitis' and 'UC' are used as the keywords, and the relevant literature is collected from online databases (Elsevier, PubMed, and Web of Science). RESULTS AND CONCLUSION Indirubin, indigo, isatin, tryptanthrin, and β-sitosterol are considered the key components in the treatment of UC with IN. Both preclinical and clinical studies support the efficacy of IN for UC, especially in severe UC or in those who do not respond to or have poor efficacy with existing therapies. The mechanisms of IN for UC are associated with the aryl hydrocarbon receptor pathway activation, immune regulation, oxidative stress inhibition, and intestinal microbial modulation. However, the clinical use of IN has the risks of adverse events such as pulmonary hypertension, which suggests the necessity for its rational application. As a potential therapeutic agent for UC that is currently receiving more attention, the clinical value of IN has been initially demonstrated and warrants further evaluation.
Collapse
Affiliation(s)
- Yu Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liu-lin Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin-zhen Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan-zhu Qian
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming-quan Wu
- Department of Pharmacy, Sichuan Provincial Orthopedic Hospital, Chengdu, China
| | - Juan Wang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai-hua Qin
- Health Preservation and Rehabilitation College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiao-bo Ye
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Gu S, Xue Y, Liu X, Tang Y, Wang D, Wu D, Yao M, Xia Z, Yang S, Cai G, Xue S, Dou D. Clinical, pharmacology and in vivo studies of QingDai (indigo naturalis) promotes mucosal healing and symptom improvement in ulcerative colitis by regulating the AHR-Th17/Treg pathway. J Inflamm (Lond) 2024; 21:44. [PMID: 39501249 PMCID: PMC11536804 DOI: 10.1186/s12950-024-00413-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/14/2024] [Indexed: 11/09/2024] Open
Abstract
Qingdai (QD), derived from various plant sources, is commonly used in traditional Chinese medicine for ulcerative colitis (UC) treatment. However, the clinical efficacy and mechanisms of orally administered QD remain unclear. This study aims to evaluate QD's efficacy in UC treatment and uncover its active components and mechanisms. A randomized controlled trial compared QD to Adisa, followed by UPLC-Q-TOF/MS and network pharmacology analyses to identify QD's core components and targets. In vivo experiments on a UC mouse model explored QD's impact on the AHR-Th17/Treg pathway using PCR, WB, ELISA, and flow cytometry. Results showed QD's efficacy in UC treatment, with mucosal healing and remission comparable to Adisa. UPLC-Q-TOF/MS identified 16 core components in mouse colon tissue, with network pharmacology revealing 67 targets, potentially involving the IL-17 signaling pathway and Th17 cell differentiation. QD and its components up-regulated AHR, CYP1A1, and Foxp3, while down-regulating RORγt. Additionally, QD modulated pro-inflammatory (IL-6, IL-17 A) and anti-inflammatory (IL-10, TGF-β1) factors, and Treg/Th17 cell ratios in LPMC. Oral QD administration effectively promoted mucosal healing and improved UC symptoms, potentially through AHR-Th17/Treg pathway regulation.
Collapse
Affiliation(s)
- Sizhen Gu
- Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yan Xue
- Department of Traditional Chinese Medicine, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Centre), China, Shanghai, 201619, P.R. China
| | - Xiaowen Liu
- Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yini Tang
- Department of Digestive Endoscopy Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Dong Wang
- Department of Gastroenterology, The second hospital of Hebei Medical University, Shijiazhuang City, 05000, Hebei Province, P.R. China
| | - Dongmei Wu
- Department of Pathology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Mingrong Yao
- Department of Radiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Zehua Xia
- Department of Anorectal, Shuguang Hospital affiliated to Shanghai University of traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Sen Yang
- Department of Clinical Lab, Shuguang Hospital affiliated to Shanghai University of traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Gan Cai
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Shigui Xue
- Department of Digestive Endoscopy Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
| | - Danbo Dou
- Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
| |
Collapse
|
5
|
Lin Y, Yang L, Wang D, Lei H, Zhang Y, Sun W, Liu J. Indigo alleviates psoriasis through the AhR/NF-κB signaling pathway: an in vitro and in vivo study. PeerJ 2024; 12:e18326. [PMID: 39465158 PMCID: PMC11505883 DOI: 10.7717/peerj.18326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Background Psoriasis is a chronic inflammatory skin disease. A strong association between the AhR/ NFκB axis and the inflammatory response in psoriasis. Indigo (IDG) has demonstrated significant anti-inflammatory properties. This study aimed to assess the anti-psoriatic efficacy of IDG while investigating the underlying mechanisms involved. Methods In the in vitro experiments, cell viability was assessed using the CCK-8. qRT-PCR was employed to measure the mRNA levels of NF-κB, TNF-α, IL-1β, AhR, and CYP1A1. Western blotting was conducted to examine alterations in cytoplasmic and nuclear AhR protein levels. Additionally, an IDG nanoemulsion (NE) cream was prepared for the in vivo experiments. A psoriasis-like skin lesion mice model was induced using IMQ (62.5 mg/day for 7 days). The severity of psoriasis was evaluated using PASI, and skin lesions were scored while epidermal thickness was assessed via HE staining. The expression of inflammatory markers, including IL-6, IL-13, IL-17A, MCP-1, and TNF-α, was detected in skin lesions using Luminex. The levels of CYP1A1, p65, and p-p65 proteins were determined by Western blotting. Results LPS stimulation significantly elevated TNF-α, IL-6, and NF-κB mRNA levels, which were notably reduced by IDG treatment. Additionally, IDG significantly enhanced the expression of AhR and CYP1A1 mRNA. Further investigation revealed that IDG facilitated AhR translocation from the cytoplasm to the nucleus. In the IMQ-induced psoriasis-like mouse model, IDG NE substantially ameliorated the severity of skin lesions. Moreover, IDG NE treatment reduced the upregulation of inflammatory cytokines such as IL-6, IL-17A, MCP-1, and TNF-α in IMQ-induced skin lesions. It was also observed that IDG NE treatment increased CYP1A1 protein expression while inhibiting p65 and p-p65 protein expression. Conclusion IDG emerges as a promising treatment for psoriasis, demonstrating effective therapeutic outcomes. Its mechanism of action is likely linked to the modulation of the AhR/NFκB signaling pathway.
Collapse
Affiliation(s)
- Yu Lin
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Dermatology, the First Affliated Hospital of Jinan University, Guangzhou, China
| | - Lihong Yang
- Department of Dermatology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Dongxiang Wang
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiqing Lei
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuelin Zhang
- Department of Dermatology, the First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen Sun
- Department of Dermatology, Jingmen Central Hospital, Jingmen, China
| | - Jing Liu
- Department of Dermatology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Zaki K, Ouabane M, Guendouzi A, Sbai A, Sekkate C, Bouachrine M, Lakhlifi T. From farm to pharma: Investigation of the therapeutic potential of the dietary plants Apium graveolens L., Coriandrum sativum, and Mentha longifolia, as AhR modulators for Immunotherapy. Comput Biol Med 2024; 181:109051. [PMID: 39186905 DOI: 10.1016/j.compbiomed.2024.109051] [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: 04/14/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Autoimmune diseases represent a complex array of conditions where the body's immune system mistakenly attacks its own tissues. These disorders, affecting millions worldwide, encompass a broad spectrum of conditions ranging from rheumatoid arthritis and multiple sclerosis to lupus and type 1 diabetes. The Aryl hydrocarbon receptor (AhR) translocator, expressed across immune and other cell types, plays crucial roles in immune disorders and inflammatory diseases. With a realm towards natural remedies in modern medicine for disease prevention, this study investigates the electronic properties and behaviors of bioactive compounds from dietary sources, including Apium graveolens L. (Celery), Coriandrum sativum seeds (Coriander), and Mentha longifolia, as AhR modulators. Through comprehensive analysis (HOMO-LUMO, ESP, LOL, and ELF), electron-rich and -poor regions, electron localization, and delocalization are identified, contrasting these compounds with the toxic AhR ligand, TCDD. Evaluation of Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) properties reveals favorable pharmacokinetics without blood-brain barrier penetration, indicating drug-like characteristics. Molecular docking demonstrates stronger interactions of dietary flavonoid ligands with AhR transcription compared to TCDD. Molecular dynamics simulations confirm the stability of complexes and the sustainability of interactions formed. This research underscores the potential of natural compounds as effective AhR modulators for therapeutic interventions in immune-related disorders.
Collapse
Affiliation(s)
- Khadija Zaki
- Molecular Chemistry and Natural Substances Laboratory, Moulay Ismail University, Faculty of Science, Meknes, Morocco
| | - Mohamed Ouabane
- Molecular Chemistry and Natural Substances Laboratory, Moulay Ismail University, Faculty of Science, Meknes, Morocco; Chemistry-Biology Applied to the Environment URL CNRT 13, Department of Chemistry, Faculty of Science, My Ismail University, Meknes, Morocco
| | - Abdelkrim Guendouzi
- Laboratory of Chemistry, Synthesis, Properties and Applications, Department of Chemistry, Faculty of Science, University of Saida, Algeria
| | - Abdelouahid Sbai
- Molecular Chemistry and Natural Substances Laboratory, Moulay Ismail University, Faculty of Science, Meknes, Morocco.
| | - Chakib Sekkate
- Chemistry-Biology Applied to the Environment URL CNRT 13, Department of Chemistry, Faculty of Science, My Ismail University, Meknes, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Moulay Ismail University, Faculty of Science, Meknes, Morocco
| | - Tahar Lakhlifi
- Molecular Chemistry and Natural Substances Laboratory, Moulay Ismail University, Faculty of Science, Meknes, Morocco
| |
Collapse
|
7
|
Wu X, Cao Y, Liu Y, Zheng J. A New Strategy for Dietary Nutrition to Improve Intestinal Homeostasis in Diarrheal Irritable Bowel Syndrome: A Perspective on Intestinal Flora and Intestinal Epithelial Interaction. Nutrients 2024; 16:3192. [PMID: 39339792 PMCID: PMC11435304 DOI: 10.3390/nu16183192] [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: 08/22/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Although a reasonable diet is essential for promoting human health, precise nutritional regulation presents a challenge for different physiological conditions. Irritable Bowel Syndrome (IBS) is characterized by recurrent abdominal pain and abnormal bowel habits, and diarrheal IBS (IBS-D) is the most common, seriously affecting patients' quality of life. Therefore, the implementation of precise nutritional interventions for IBS-D has become an urgent challenge in the fields of nutrition and food science. IBS-D intestinal homeostatic imbalance involves intestinal flora disorganization and impaired intestinal epithelial barrier function. A familiar interaction is evident between intestinal flora and intestinal epithelial cells (IECs), which together maintain intestinal homeostasis and health. Dietary patterns, such as the Mediterranean diet, have been shown to regulate gut flora, which in turn improves the body's health by influencing the immune system, the hormonal system, and other metabolic pathways. METHODS This review summarized the relationship between intestinal flora, IECs, and IBS-D. It analyzed the mechanism behind IBS-D intestinal homeostatic imbalance by examining the interactions between intestinal flora and IECs, and proposed a precise dietary nutrient intervention strategy. RESULTS AND CONCLUSION This increases the understanding of the IBS-D-targeted regulation pathways and provides guidance for designing related nutritional intervention strategies.
Collapse
Affiliation(s)
- Xinyu Wu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (X.W.); (Y.C.)
| | - Yilong Cao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (X.W.); (Y.C.)
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (X.W.); (Y.C.)
| | - Jie Zheng
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
8
|
Shimada F, Yoshimatsu Y, Sujino T, Fukuda T, Aoki Y, Hayashi Y, Tojo A, Kawaguchi T, Kiyohara H, Sugimoto S, Nanki K, Mikami Y, Miyamoto K, Takabayashi K, Hosoe N, Kato M, Ogata H, Naganuma M, Kanai T. Clinical outcomes of patients with remitting ulcerative colitis after discontinuation of indigo naturalis. Sci Rep 2024; 14:5778. [PMID: 38459203 PMCID: PMC10923923 DOI: 10.1038/s41598-024-56543-y] [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: 12/19/2023] [Accepted: 03/07/2024] [Indexed: 03/10/2024] Open
Abstract
Indigo naturalis is an effective treatment for ulcerative colitis. However, long-term use of indigo naturalis causes adverse events, such as pulmonary hypertension. The natural history of patients with ulcerative colitis who discontinued indigo naturalis after induction therapy is unknown. Moreover, the clinical features of patients who relapsed within 52 weeks after the discontinuation of indigo naturalis are unclear. This study aimed to assess the clinical outcomes of patients with ulcerative colitis after discontinuation of indigo naturalis and to identify potential markers responsible for relapse. This single-center retrospective study investigated the follow-up of 72 patients who achieved a clinical response 8 weeks after indigo naturalis treatment. We observed relapse in patients with ulcerative colitis after the discontinuation of indigo naturalis. We analyzed the factors predicting long-term outcomes after discontinuation of indigo naturalis. Relapse was observed in 24%, 57%, and 71% of patients at 8, 26, and 52 weeks, respectively. There were no predictive markers in patients who relapsed within 52 weeks after the discontinuation of indigo naturalis. The ulcerative colitis relapse rate after indigo naturalis discontinuation was high. Follow-up treatment is required after the discontinuation of indigo naturalis in patients with ulcerative colitis.
Collapse
Affiliation(s)
- Fumie Shimada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yusuke Yoshimatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Tomohisa Sujino
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Tomohiro Fukuda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Division of Gastroenterology, Yokohama Municipal Citizen's Hospital, 1-1, Nishimachi, Mitsuzawa, Kanagawaku, Yokohama, Kanagawa, 221-0855, Japan
| | - Yasuhiro Aoki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yukie Hayashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Anna Tojo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takaaki Kawaguchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroki Kiyohara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shinya Sugimoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kosaku Nanki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kentaro Miyamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Miyarisan Pharmaceutical Co., Ltd., 1-10-3, Kaminakazato, Kita-ku, Tokyo, 114-0016, Japan
| | - Kaoru Takabayashi
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naoki Hosoe
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Motohiko Kato
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Haruhiko Ogata
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Makoto Naganuma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-3-1, Shinmachi, Maikatashi, Osaka, 573-1191, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
9
|
Rayan M, Sayed TS, Hussein OJ, Therachiyil L, Maayah ZH, Maccalli C, Uddin S, Prehn JHM, Korashy HM. Unlocking the secrets: exploring the influence of the aryl hydrocarbon receptor and microbiome on cancer development. Cell Mol Biol Lett 2024; 29:33. [PMID: 38448800 PMCID: PMC10918910 DOI: 10.1186/s11658-024-00538-0] [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: 09/11/2023] [Accepted: 01/17/2024] [Indexed: 03/08/2024] Open
Abstract
Gut microbiota regulates various aspects of human physiology by producing metabolites, metabolizing enzymes, and toxins. Many studies have linked microbiota with human health and altered microbiome configurations with the occurrence of several diseases, including cancer. Accumulating evidence suggests that the microbiome can influence the initiation and progression of several cancers. Moreover, some microbiotas of the gut and oral cavity have been reported to infect tumors, initiate metastasis, and promote the spread of cancer to distant organs, thereby influencing the clinical outcome of cancer patients. The gut microbiome has recently been reported to interact with environmental factors such as diet and exposure to environmental toxicants. Exposure to environmental pollutants such as polycyclic aromatic hydrocarbons (PAHs) induces a shift in the gut microbiome metabolic pathways, favoring a proinflammatory microenvironment. In addition, other studies have also correlated cancer incidence with exposure to PAHs. PAHs are known to induce organ carcinogenesis through activating a ligand-activated transcriptional factor termed the aryl hydrocarbon receptor (AhR), which metabolizes PAHs to highly reactive carcinogenic intermediates. However, the crosstalk between AhR and the microbiome in mediating carcinogenesis is poorly reviewed. This review aims to discuss the role of exposure to environmental pollutants and activation of AhR on microbiome-associated cancer progression and explore the underlying molecular mechanisms involved in cancer development.
Collapse
Affiliation(s)
- Menatallah Rayan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Tahseen S Sayed
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Ola J Hussein
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | - Lubna Therachiyil
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Zaid H Maayah
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar
| | | | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, P. O. Box 2713, Doha, Qatar.
| |
Collapse
|
10
|
Xu Y, Lin C, Tan HY, Bian ZX. The double-edged sword effect of indigo naturalis. Food Chem Toxicol 2024; 185:114476. [PMID: 38301993 DOI: 10.1016/j.fct.2024.114476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/03/2024]
Abstract
Indigo naturalis (IN) is a dried powder derived from plants such as Baphicacanthus cusia (Neeks) Bremek., Polygonum tinctorium Ait. and Isatis indigotica Fork. It has a historical application as a dye in ancient India, Egypt, Africa and China. Over time, it has been introduced to China and Japan for treatment of various ailments including hemoptysis, epistaxis, chest discomfort, and aphtha. Clinical and pre-clinical studies have widely demonstrated its promising effects on autoimmune diseases like psoriasis and Ulcerative colitis (UC). Despite the documented efficacy of IN in UC patients, concerns have been raised on the development of adverse effects with long term consumption, prompting a closer examination of its safety and tolerability in these contexts. This review aims to comprehensively assess the efficacy of IN in both clinical and pre-clinical settings, with a detailed exploration of the mechanisms of action involved. Additionally, it summarizes the observed potential toxicity of IN in animal and human settings was summarized. This review will deepen our understanding on the beneficial and detrimental effects of IN in UC, providing valuable insights for its future application in patients with this condition.
Collapse
Affiliation(s)
- Yiqi Xu
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chengyuan Lin
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hor-Yue Tan
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Zhao-Xiang Bian
- Centre for Chinese Herbal Medicine Drug Development, Hong Kong Baptist University, Hong Kong SAR, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
11
|
Xu X, Taha R, Chu C, Xiao L, Wang T, Wang X, Huang X, Jiang Z, Sun L. Indirubin mediates adverse intestinal reactions in guinea pigs by downregulating the expression of AchE through AhR. Xenobiotica 2024; 54:83-94. [PMID: 38164702 DOI: 10.1080/00498254.2023.2297745] [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: 10/22/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Indirubin is the main component of the traditional Chinese medicine Indigo naturalis (IN), a potent agonist of aryl hydrocarbon receptors (AhRs). In China, IN is used to treat psoriasis and ulcerative colitis, and indirubin is used for the treatment of chronic myelogenous leukaemia. However, IN and indirubin have adverse reactions, such as abdominal pain, diarrhoea, and intussusception, and their specific mechanism is unclear.The purpose of our research was to determine the specific mechanism underlying the adverse effects of IN and indirubin. By tracking the modifications in guinea pigs after the intragastric administration of indirubin for 28 days.The results demonstrate that indirubin could accelerate bowel movements and decrease intestinal acetylcholinesterase (AchE) expression. Experiments with NCM460 cells revealed that indirubin significantly reduced the expression of AchE, and the AchE levels were increased after the silencing of AhR and re-exposure to indirubin.This study showed that the inhibition of AchE expression by indirubin plays a key role in the occurrence of adverse reactions to indirubin and that the underlying mechanism is related to AhR-mediated AchE downregulation.
Collapse
Affiliation(s)
- Xiaoting Xu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Reham Taha
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Chenghan Chu
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Li Xiao
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, China
| | - Tao Wang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Xinzhi Wang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Xin Huang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Zhenzhou Jiang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Lixin Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
12
|
Ben-Horin S, Salomon N, Karampekos G, Viazis N, Lahat A, Ungar B, Eliakim R, Kuperstein R, Kriger-Sharabi O, Reiss-Mintz H, Yanai H, Dotan I, Zittan E, Maharshak N, Hirsch A, Weitman M, Mantzaris GJ, Kopylov U. Curcumin-QingDai Combination for Patients With Active Ulcerative Colitis: A Randomized, Double-Blinded, Placebo-Controlled Trial. Clin Gastroenterol Hepatol 2024; 22:347-356.e6. [PMID: 37302449 DOI: 10.1016/j.cgh.2023.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND & AIMS We evaluated the efficacy of herbal combination of curcumin-QingDai (CurQD) in active ulcerative colitis (UC). METHODS Part I was an open-label trial of CurQD in patients with active UC, defined by a Simple Clinical Colitis Activity Index score of 5 or higher and a Mayo endoscopic subscore of 2 or higher. Part II was a placebo-controlled trial conducted in Israel and Greece, randomizing active UC patients at a 2:1 ratio to enteric-coated CurQD 3 g/d or placebo for 8 weeks. The co-primary outcome was clinical response (reduction in the Simple Clinical Colitis Activity Index of ≥3 points) and an objective response (Mayo endoscopic subscore improvement of ≥1 or a 50% fecal calprotectin reduction). Responding patients continued either maintenance curcumin or placebo alone for an additional 8 weeks. Aryl-hydrocarbon receptor activation was assessed by cytochrome P450 1A1 (CYP1A1) mucosal expression. RESULTS In part I, 7 of 10 patients responded and 3 of 10 achieved clinical remission. Of 42 patients in part II, the week 8 co-primary outcome was achieved in 43% and 8% of CurQD and placebo patients, respectively (P = .033). Clinical response was observed in 85.7% vs 30.7% (P < .001), clinical remission in 14 of 28 (50%) vs 1 of 13 (8%; P = .01), a 50% calprotectin reduction in 46.4% vs 15.4% (P = .08), and endoscopic improvement in 75% vs 20% (P = .036) in the CurQD and placebo groups, respectively. Adverse events were comparable between groups. By week 16, curcumin-maintained clinical response, clinical remission, and clinical biomarker response rates were 93%, 80%, and 40%, respectively. CurQD uniquely up-regulated mucosal CYP1A1 expression, which was not observed among patients receiving placebo, mesalamine, or biologics. CONCLUSIONS In this placebo-controlled trial, CurQD was effective for inducing response and remission in active UC patients. The aryl-hydrocarbon receptor pathway may merit further study as a potential UC treatment target. CLINICALTRIALS gov ID: NCT03720002.
Collapse
Affiliation(s)
- Shomron Ben-Horin
- Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel; School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Nir Salomon
- Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel.
| | - Georgios Karampekos
- Department of Gastroenterology, Evangelismos-Polykliniki General Hospital of Athens, Athens, Greece
| | - Nikos Viazis
- Department of Gastroenterology, Evangelismos-Polykliniki General Hospital of Athens, Athens, Greece
| | - Adi Lahat
- Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel; School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Bella Ungar
- Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel; School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rami Eliakim
- Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel; School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rafael Kuperstein
- School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Leviev Heart Center, Sheba Medical Center, Ramat-Gan, Israel
| | | | - Hilla Reiss-Mintz
- Gastroenterology Department, Mayanei HaYeshua Medical Center, Bnei Brak, Israel
| | - Henit Yanai
- School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
| | - Iris Dotan
- School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
| | - Eran Zittan
- Department of Gastroenterology and Liver Diseases, Emek Medical Center, Afula, Israel; The Rappaport Faculty of Medicine Technion, Israel Institute of Technology, Haifa, Israel
| | - Nitsan Maharshak
- School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Department of Gastroenterology, Tel Aviv Medical Center, Tel-Aviv, Israel
| | - Ayal Hirsch
- School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Department of Gastroenterology, Tel Aviv Medical Center, Tel-Aviv, Israel
| | - Michal Weitman
- MS Unit, Chemistry Department, Bar llan University, Ramat-Gan, Israel
| | - Gerassimos J Mantzaris
- Department of Gastroenterology, Evangelismos-Polykliniki General Hospital of Athens, Athens, Greece
| | - Uri Kopylov
- Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel; School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
13
|
Yao Y, Liu Y, Xu Q, Mao L. Short Chain Fatty Acids: Essential Weapons of Traditional Medicine in Treating Inflammatory Bowel Disease. Molecules 2024; 29:379. [PMID: 38257292 PMCID: PMC10818876 DOI: 10.3390/molecules29020379] [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: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and recurrent intestinal inflammatory disease, mainly including Crohn's disease (CD) and ulcerative colitis (UC). In recent years, the incidence and prevalence of IBD have been on the rise worldwide and have become a significant concern of health and a huge economic burden on patients. The occurrence and development of IBD involve a variety of pathogenic factors. The changes in short-chain fatty acids (SCFAs) are considered to be an important pathogenic mechanism of this disease. SCFAs are important metabolites in the intestinal microbial environment, which are closely involved in regulating immune, anti-tumor, and anti-inflammatory activities. Changes in metabolite levels can reflect the homeostasis of the intestinal microflora. Recent studies have shown that SCFAs provide energy for host cells and intestinal microflora, shape the intestinal environment, and regulate the immune system, thereby regulating intestinal physiology. SCFAs can effectively reduce the incidence of enteritis, cardiovascular disease, colon cancer, obesity, and diabetes, and also play an important role in maintaining the balance of energy metabolism (mainly glucose metabolism) and improving insulin tolerance. In recent years, many studies have shown that numerous decoctions and natural compounds of traditional Chinese medicine have shown promising therapeutic activities in multiple animal models of colitis and thus attracted increasing attention from scientists in the study of IBD treatment. Some of these traditional Chinese medicines or compounds can effectively alleviate colonic inflammation and clinical symptoms by regulating the generation of SCFAs. This study reviews the effects of various traditional Chinese medicines or bioactive substances on the production of SCFAs and their potential impacts on the severity of colonic inflammation. On this basis, we discussed the mechanism of SCFAs in regulating IBD-associated inflammation, as well as the related regulatory factors and signaling pathways. In addition, we provide our understanding of the limitations of current research and the prospects for future studies on the development of new IBD therapies by targeting SCFAs. This review may widen our understanding of the effect of traditional medicine from the view of SCFAs and their role in alleviating IBD animal models, thus contributing to the studies of IBD researchers.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| | - Yongchao Liu
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| | - Qiuyun Xu
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| |
Collapse
|
14
|
Chandel N, Singh BB, Dureja C, Yang YH, Bhatia SK. Indigo production goes green: a review on opportunities and challenges of fermentative production. World J Microbiol Biotechnol 2024; 40:62. [PMID: 38182914 DOI: 10.1007/s11274-023-03871-2] [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: 10/11/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024]
Abstract
Indigo is a widely used dye in various industries, such as textile, cosmetics, and food. However, traditional methods of indigo extraction and processing are associated with environmental and economic challenges. Fermentative production of indigo using microbial strains has emerged as a promising alternative that offers sustainability and cost-effectiveness. This review article provides a critical overview of microbial diversity, metabolic pathways, fermentation strategies, and genetic engineering approaches for fermentative indigo production. The advantages and limitations of different indigo production systems and a critique of the current understanding of indigo biosynthesis are discussed. Finally, the potential application of indigo in other sectors is also discussed. Overall, fermentative production of indigo offers a sustainable and bio-based alternative to synthetic methods and has the potential to contribute to the development of sustainable and circular biomanufacturing.
Collapse
Affiliation(s)
- Neha Chandel
- School of Medical and Allied Sciences, GD Goenka University, Gurugram, Haryana, 122103, India
| | - Bharat Bhushan Singh
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chetna Dureja
- Center for Inflammatory and Infectious Diseases, Texas A&M Health Science Center, Institute of Bioscience and Technology, Houston, TX, USA
| | - Yung-Hun Yang
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea
- Institute for Ubiquitous Information Technology and Applications, Seoul, 05029, Republic of Korea
| | - Shashi Kant Bhatia
- Department of Biological Engineering, College of Engineering, Konkuk University, Seoul, 05029, Republic of Korea.
- Institute for Ubiquitous Information Technology and Applications, Seoul, 05029, Republic of Korea.
| |
Collapse
|
15
|
Yao Y, Li X, Yang X, Mou H, Wei L. Indirubin, an Active Component of Indigo Naturalis, Exhibits Inhibitory Effects on Leukemia Cells via Targeting HSP90AA1 and PI3K/Akt Pathway. Anticancer Agents Med Chem 2024; 24:718-727. [PMID: 38347773 DOI: 10.2174/0118715206258293231017063340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/10/2023] [Accepted: 09/14/2023] [Indexed: 06/04/2024]
Abstract
BACKGROUND This research intended to predict the active ingredients and key target genes of Indigo Naturalis in treating human chronic myeloid leukemia (CML) using network pharmacology and conduct the invitro verification. METHODS The active components of Indigo Naturalis and the corresponding targets and leukemia-associated genes were gathered through public databases. The core targets and pathways of Indigo Naturalis were predicted through protein-protein interaction (PPI) network, gene ontology (GO) function, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Next, after intersecting with leukemia-related genes, the direct core target gene of Indigo Naturalis active components was identified. Subsequently, HL-60 cells were stimulated with indirubin (IND) and then examined for cell proliferation using CCK-8 assay and cell cycle, cell apoptosis, and mitochondrial membrane potential using flow cytometry. The content of apoptosis-associated proteins (Cleaved Caspase 9, Cleaved Caspase 7, Cleaved Caspase 3, and Cleaved parp) were detected using Western blot, HSP90AA1 protein, and PI3K/Akt signaling (PI3K, p-PI3K, Akt, and p-Akt) within HL-60 cells. RESULTS A total of 9 active components of Indigo Naturalis were screened. The top 10 core target genes (TNF, PTGS2, RELA, MAPK14, IFNG, PPARG, NOS2, IKBKB, HSP90AA1, and NOS3) of Indigo Naturalis active components within the PPI network were identified. According to the KEGG enrichment analysis, these targets were associated with leukemia-related pathways (such as acute myeloid leukemia and CML). After intersecting with leukemia-related genes, it was found that IND participated in the most pairs of target information and was at the core of the target network; HSP90AA1 was the direct core gene of IND. Furthermore, the in-vitro cell experiments verified that IND could inhibit the proliferation, elicit G2/M-phase cell cycle arrest, enhance the apoptosis of HL-60 cells, reduce mitochondrial membrane potential, and promote apoptosis-related protein levels. Under IND treatment, HSP90AA1 overexpression notably promoted cell proliferation and inhibited apoptosis. Additionally, IND exerted tumor suppressor effects on leukemia cells by inhibiting HSP90AA1 expression. CONCLUSION IND, an active component of Indigo Naturalis, could inhibit CML progression, which may be achieved via inhibiting HSP90AA1 and PI3K/Akt signaling expression levels.
Collapse
MESH Headings
- Humans
- Cell Proliferation/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Apoptosis/drug effects
- Phosphatidylinositol 3-Kinases/metabolism
- Indoles/pharmacology
- Indoles/chemistry
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Drug Screening Assays, Antitumor
- HL-60 Cells
- Molecular Structure
- Dose-Response Relationship, Drug
- Structure-Activity Relationship
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia/drug therapy
- Leukemia/pathology
- Leukemia/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Yuanzhi Yao
- College of Biology and Food Engineering, Huaihua University, Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Huaihua 418000, China
| | - Xiaoying Li
- College of Biology and Food Engineering, Huaihua University, Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Huaihua 418000, China
| | - Xiaoqin Yang
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Hai Mou
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Lin Wei
- College of Biology and Food Engineering, Huaihua University, Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Huaihua 418000, China
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| |
Collapse
|
16
|
Xie J, Huang Q, Xie H, Liu J, Tian S, Cao R, Yang M, Lin J, Han L, Zhang D. Hyaluronic acid/inulin-based nanocrystals with an optimized ratio of indigo and indirubin for combined ulcerative colitis therapy via immune and intestinal flora regulation. Int J Biol Macromol 2023; 252:126502. [PMID: 37625742 DOI: 10.1016/j.ijbiomac.2023.126502] [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: 04/18/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Indigo (IND) and indirubin (INB) have demonstrated a synergistic effect in treating ulcerative colitis at a ratio of 7.5:1. However, the colon mucus layer, a critical physiological barrier against external threats, is also a biological barrier, limiting the potential for effective drug delivery to the lamina propria for regulating inflammatory cells. Inspired by the potential of Hyaluronic acid (HA), to enhance cellular uptake by inflammatory cells, and Pluronic® F127 (F127), known for overcoming the mucus barrier, this study innovatively developed INB/IND nanosuspensions by co-modifying with F127 and HA. Moreover, inulin serves a dual purpose as a spray protective agent and a regulator of intestinal flora. Therefore, it was incorporated into INB/IND nanosuspensions for subsequent spray drying, resulting in the preparation of INB/IND nanocrystals (INB/IND-NC). The mucus penetration of INB/IND-NC was 24.30 times that of the control group. Besides, INB/IND-NC exhibited enhanced cellular uptake properties proximately twice that of Raw INB/IND. Importantly, INB/IND-NC exhibited improved therapeutic efficacy in DSS-induced mice by regulating the expression of cytokines, regulating immune responses via downregulating the expression of macrophages, neutrophils, and dendritic cells and maintaining intestinal flora homeostasis. Our study provides a new perspective for applying natural products for treating inflammatory diseases.
Collapse
Affiliation(s)
- Jin Xie
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qi Huang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huijuan Xie
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jun Liu
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shimin Tian
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ruiyi Cao
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Junzhi Lin
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Li Han
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dingkun Zhang
- State Key Laboratory of Characteristic Chinese Drug Resources in Southwest China, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
17
|
Zhu MZ, Yang MF, Song Y, Xu HM, Xu J, Yue NN, Zhang Y, Tian CM, Shi RY, Liang YJ, Yao J, Wang LS, Nie YQ, Li DF. Exploring the efficacy of herbal medicinal products as oral therapy for inflammatory bowel disease. Biomed Pharmacother 2023; 165:115266. [PMID: 37541177 DOI: 10.1016/j.biopha.2023.115266] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) encompasses a collection of idiopathic diseases characterized by chronic inflammation in the gastrointestinal (GI) tract. Patients diagnosed with IBD often experience necessitate long-term pharmacological interventions. Among the multitude of administration routes available for treating IBD, oral administration has gained significant popularity owing to its convenience and widespread utilization. In recent years, there has been extensive evaluation of the efficacy of orally administered herbal medicinal products and their extracts as a means of treating IBD. Consequently, substantial evidence has emerged, supporting their effectiveness in IBD treatment. This review aimed to provide a comprehensive summary of recent studies evaluating the effects of herbal medicinal products in the treatment of IBD. We delved into the regulatory role of these products in modulating immunity and maintaining the integrity of the intestinal epithelial barrier. Additionally, we examined their impact on antioxidant activity, anti-inflammatory properties, and the modulation of intestinal flora. By exploring these aspects, we aimed to emphasize the significant advantages associated with the use of oral herbal medicinal products in the treatment of IBD. Of particular note, this review introduced the concept of herbal plant-derived exosome-like nanoparticles (PDENs) as the active ingredient in herbal medicinal products for the treatment of IBD. The inclusion of PDENs offers distinct advantages, including enhanced tissue penetration and improved physical and chemical stability. These unique attributes not only demonstrate the potential of PDENs but also pave the way for the modernization of herbal medicinal products in IBD treatment.
Collapse
Affiliation(s)
- Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen 518020, Guangdong, China
| | - Yang Song
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University), Shenzhen 518020, Guangdong, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou 516000, Guangdong, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Rui-Yue Shi
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen 518020, Guangdong, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China.
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China.
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University; the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
18
|
Yokote A, Imazu N, Umeno J, Kawasaki K, Fujioka S, Fuyuno Y, Matsuno Y, Moriyama T, Miyawaki K, Akashi K, Kitazono T, Torisu T. Ferroptosis in the colon epithelial cells as a therapeutic target for ulcerative colitis. J Gastroenterol 2023; 58:868-882. [PMID: 37410250 DOI: 10.1007/s00535-023-02016-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Ferroptosis, a type of programmed cell death triggered by oxidative stress, was suspected to play a role in ulcerative colitis. Indigo naturalis is highly effective against ulcerative colitis, but its mechanism is unclear. This study found that indigo naturalis treatment suppressed ferroptosis. METHODS We analyzed 770 mRNA expressions of patients with ulcerative colitis. Suppression of ferroptosis by indigo naturalis treatment was shown using a cell death assay. Malondialdehyde levels and reactive oxygen species were analyzed in CaCo-2 cells treated with indigo naturalis. Glutathione metabolism was shown by metabolomic analysis. Extraction of the ingredients indigo naturalis from the rectal mucosa was performed using liquid chromatograph-mass spectrometry. RESULTS Gene expression profiling showed that indigo naturalis treatment increased antioxidant genes in the mucosa of patients with ulcerative colitis. In vitro analysis showed that nuclear factor erythroid-2-related factor 2-related antioxidant gene expression was upregulated by indigo naturalis. Indigo naturalis treatment rendered cells resistant to ferroptosis. Metabolomic analysis suggested that an increase in reduced glutathione by indigo naturalis. The protein expression of CYP1A1 and GPX4 was increased in the rectum by treatment with indigo naturalis. The main ingredients of indigo naturalis, indirubin and indigo inhibited ferroptosis. Indirubin was detected in the rectal mucosa of patients with ulcerative colitis who were treated with indigo naturalis. CONCLUSIONS Suppression of ferroptosis by indigo naturalis in the intestinal epithelium could be therapeutic target for ulcerative colitis. The main active ingredient of indigo naturalis may be indirubin.
Collapse
Affiliation(s)
- Akihito Yokote
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Noriyuki Imazu
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Junji Umeno
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Keisuke Kawasaki
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shin Fujioka
- Department of Endoscopic Diagnostics and Therapeutics, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Yuta Fuyuno
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuichi Matsuno
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiko Moriyama
- International Medical Department, Kyushu University Hospital, Fukuoka, 812-8582, Japan
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takehiro Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
19
|
Shan J, Liu S, Liu H, Yuan J, Lin J. Mechanism of Qingchang Suppository on repairing the intestinal mucosal barrier in ulcerative colitis. Front Pharmacol 2023; 14:1221849. [PMID: 37675045 PMCID: PMC10478270 DOI: 10.3389/fphar.2023.1221849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Ulcerative colitis (UC) is a refractory inflammatory bowel disease, and the outcomes of conventional therapies of UC, including 5-aminosalicylic acid, glucocorticoids, immunosuppressants, and biological agents, are not satisfied with patients and physicians with regard to adverse reactions and financial burden. The abnormality of the intestinal mucosal barrier in the pathogenesis of UC was verified. Qingchang Suppository (QCS) is an herbal preparation and is effective in treating ulcerative proctitis. The mechanism of QCS and its active ingredients have not been concluded especially in mucosal healing. This review elucidated the potential mechanism of QCS from the intestinal mucosal barrier perspective to help exploring future QCS research directions.
Collapse
Affiliation(s)
- Jingyi Shan
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Suxian Liu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haoyue Liu
- Department of Intensive Care Unit, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianye Yuan
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiang Lin
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Yanai H, Salomon N, Lahat A, Ungar B, Eliakim R, Kriger-Sharabi O, Reiss-Mintz H, Koslowsky B, Shitrit ABG, Tamir-Degabli N, Dotan I, Zittan E, Maharshak N, Hirsch A, Ben-Horin S, Kopylov U. Real-world experience with Curcumin-QingDai combination for patients with active ulcerative colitis: A retrospective multicentre cohort study. Aliment Pharmacol Ther 2023; 58:175-181. [PMID: 37157131 DOI: 10.1111/apt.17538] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/06/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Curcumin and QingDai (QD, Indigo) have been shown to be effective for treating active ulcerative colitis (UC). AIM To evaluate the real-world experience with the Curcumin-QingDai (CurQD) herbal combination to induce remission in active UC. METHODS A retrospec-tive multicentre adult cohort study from five tertiary academic centres (2018-2022). Active UC was defined as a Simple Clinical Colitis Activity Index (SCCAI) ≥ 3. Patients were induced by CurQD. The primary outcome was clinical remission at weeks 8-12, defined as SCCAI ≤2 and a decrease ≥3 points from baseline. Secondary outcomes were clinical response (SCCAI decrease ≥3 points), corticosteroid-free remission, faecal calprotectin (FC) response (reduction ≥50%), FC normalisation (FC ≤100 μg/g for patients with FC ≥300 μg/g at baseline), and safety. All outcomes were analysed for patients who were maintaining stable treatment. RESULTS Eighty-eight patients were included; 50% were biologics/small molecules experienced, and 36.5% received ≥2 biologics/small molecules. Clinical remission was achieved in 41 (46.5%), and clinical response in 53 (60.2%). Median SCCAI decreased from 7 (IQR:5-9) to 2 (IQR:1-3); p < 0.0001. Of the 26 patients on corticosteroids at baseline, seven achieved corticosteroid-free remission. Among 43 biologics/small molecules experienced patients, clinical remission was achieved in 39.5% and clinical response in 58.1%. FC normalisation and response were achieved in 17/29 and 27/33, respectively. Median FC decreased from 1000 μg/g (IQR:392-2772) at baseline to 75 μg/g (IQR:12-136) at the end of inductions (n = 30 patients with paired samples); p < 0.0001. No overt safety signals emerged. CONCLUSION In this real-world cohort, CurQD effectively induced clinical and biomarker remission in patients with active UC, including patients who were biologics/small molecules experienced.
Collapse
Affiliation(s)
- Henit Yanai
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Nir Salomon
- Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
| | - Adi Lahat
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
| | - Bella Ungar
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
| | - Rami Eliakim
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
| | | | - Hilla Reiss-Mintz
- Department of Gastroenterology, Mayanei HaYeshua Medical Center, Bnei Brak, Israel
| | - Benjamin Koslowsky
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ariella Bar-Gil Shitrit
- Digestive Diseases Institute, Shaare Zedek Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Natalie Tamir-Degabli
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Internal Medicine E, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Zittan
- Department of Gastroenterology and Liver Diseases, Emek Medical Center, Afula, Israel
- The Rappaport Faculty of Medicine Technion-Israel Institute of Technology, Haifa, Israel
| | - Nitsan Maharshak
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Tel Aviv Medical Center, Tel-Aviv, Israel
| | - Ayal Hirsch
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Tel Aviv Medical Center, Tel-Aviv, Israel
| | - Shomron Ben-Horin
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
| | - Uri Kopylov
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
21
|
Huang W, Rui K, Wang X, Peng N, Zhou W, Shi X, Lu L, Hu D, Tian J. The aryl hydrocarbon receptor in immune regulation and autoimmune pathogenesis. J Autoimmun 2023; 138:103049. [PMID: 37229809 DOI: 10.1016/j.jaut.2023.103049] [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: 02/20/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023]
Abstract
As a ligand-activated transcription factor, the aryl hydrocarbon receptor (AhR) is activated by structurally diverse ligands derived from the environment, diet, microorganisms, and metabolic activity. Recent studies have demonstrated that AhR plays a key role in modulating both innate and adaptive immune responses. Moreover, AhR regulates innate immune and lymphoid cell differentiation and function, which is involved in autoimmune pathogenesis. In this review, we discuss recent advances in understanding the mechanism of activation of AhR and its mediated functional regulation in various innate immune and lymphoid cell populations, as well as the immune-regulatory effect of AhR in the development of autoimmune diseases. In addition, we highlight the identification of AhR agonists and antagonists that may serve as potential therapeutic targets for the treatment of autoimmune disorders.
Collapse
Affiliation(s)
- Wei Huang
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Xiaomeng Wang
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Na Peng
- Department of Rheumatology and Nephrology, The Second People's Hospital, China Three Gorges University, Yichang, China
| | - Wenhao Zhou
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital and School of Medicine, Henan University of Science and Technology, Luoyang, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Chongqing International Institute for Immunology, China
| | - Dajun Hu
- Department of Rheumatology and Nephrology, The Second People's Hospital, China Three Gorges University, Yichang, China.
| | - Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
22
|
Chen Y, Wang Y, Fu Y, Yin Y, Xu K. Modulating AHR function offers exciting therapeutic potential in gut immunity and inflammation. Cell Biosci 2023; 13:85. [PMID: 37179416 PMCID: PMC10182712 DOI: 10.1186/s13578-023-01046-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a classical exogenous synthetic ligand of AHR that has significant immunotoxic effects. Activation of AHR has beneficial effects on intestinal immune responses, but inactivation or overactivation of AHR can lead to intestinal immune dysregulation and even intestinal diseases. Sustained potent activation of AHR by TCDD results in impairment of the intestinal epithelial barrier. However, currently, AHR research has been more focused on elucidating physiologic AHR function than on dioxin toxicity. The appropriate level of AHR activation plays a role in maintaining gut health and protecting against intestinal inflammation. Therefore, AHR offers a crucial target to modulate intestinal immunity and inflammation. Herein, we summarize our current understanding of the relationship between AHR and intestinal immunity, the ways in which AHR affects intestinal immunity and inflammation, the effects of AHR activity on intestinal immunity and inflammation, and the effect of dietary habits on intestinal health through AHR. Finally, we discuss the therapeutic role of AHR in maintaining gut homeostasis and relieving inflammation.
Collapse
Affiliation(s)
- Yue Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Yadong Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Yawei Fu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Kang Xu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
23
|
Yoshimatsu Y, Sujino T, Kanai T. Reviewing not Homer's Iliad, but "Kai Bao Ben Cao": indigo dye-the past, present, and future. Intest Res 2023; 21:174-176. [PMID: 35692192 PMCID: PMC10169512 DOI: 10.5217/ir.2022.00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/19/2022] [Accepted: 04/23/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yusuke Yoshimatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tomohisa Sujino
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Kubota K, Imai Y, Okuyama T, Ishiyama Y, Ueno S, Kario K. Dramatically Improved Severe Pulmonary Arterial Hypertension Caused by Qing-Dai (Chinese Herbal Drug) for Ulcerative Colitis. Int Heart J 2023; 64:316-320. [PMID: 37005323 DOI: 10.1536/ihj.22-563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and fatal disease for which some causative drugs have been developed. Qing-Dai is a Chinese herbal drug that is sometimes used as a specific treatment for ulcerative colitis in Asia, including Japan. Here, we report a case of severe Qing-Dai-induced PAH. A 19-year-old woman who has been taking Qing-Dai for 8 months was admitted for exertional dyspnea. Her mean pulmonary artery pressure dramatically improved from 72 to 18 mmHg with Qing-Dai discontinuation and PAH-specific therapy. After 6 years of onset, she had not relapsed with PAH with PAH-specific therapy.
Collapse
Affiliation(s)
- Kana Kubota
- Division of Cardiovascular Medicine, Department of Internal Medicine, Jichi Medical University
| | - Yasushi Imai
- Division of Cardiovascular Medicine, Department of Internal Medicine, Jichi Medical University
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University
| | - Takafumi Okuyama
- Division of Cardiovascular Medicine, Department of Internal Medicine, Jichi Medical University
| | - Yusuke Ishiyama
- Division of Cardiovascular Medicine, Department of Internal Medicine, Jichi Medical University
| | | | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Internal Medicine, Jichi Medical University
| |
Collapse
|
25
|
Luo J, Zhang X, Wang X, Pei J, Zhao L. Directional preparation of indigo or indirubin from indican by an alkali-resistant glucosidase under specific pH and temperature. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
26
|
Asari T, Kikuchi H, Kawaguchi S, Sakuraba H, Yoshida S, Akemoto Y, Maeda T, Shinji O, Murai Y, Higuchi N, Hoshi K, Fukutoku Y, Hiraga H, Sasaki K, Fukuda S. Polygonum tinctorium leaves suppress sodium dextran sulfate-induced colitis through interleukin-10-related pathway. Biochem Biophys Rep 2022; 30:101272. [PMID: 35535330 PMCID: PMC9077533 DOI: 10.1016/j.bbrep.2022.101272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/03/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023] Open
|
27
|
Current Therapeutic Landscape and Safety Roadmap for Targeting the Aryl Hydrocarbon Receptor in Inflammatory Gastrointestinal Indications. Cells 2022; 11:cells11101708. [PMID: 35626744 PMCID: PMC9139855 DOI: 10.3390/cells11101708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/30/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
Target modulation of the AhR for inflammatory gastrointestinal (GI) conditions holds great promise but also the potential for safety liabilities both within and beyond the GI tract. The ubiquitous expression of the AhR across mammalian tissues coupled with its role in diverse signaling pathways makes development of a “clean” AhR therapeutically challenging. Ligand promiscuity and diversity in context-specific AhR activation further complicates targeting the AhR for drug development due to limitations surrounding clinical translatability. Despite these concerns, several approaches to target the AhR have been explored such as small molecules, microbials, PROTACs, and oligonucleotide-based approaches. These various chemical modalities are not without safety liabilities and require unique de-risking strategies to parse out toxicities. Collectively, these programs can benefit from in silico and in vitro methodologies that investigate specific AhR pathway activation and have the potential to implement thresholding parameters to categorize AhR ligands as “high” or “low” risk for sustained AhR activation. Exploration into transcriptomic signatures for AhR safety assessment, incorporation of physiologically-relevant in vitro model systems, and investigation into chronic activation of the AhR by structurally diverse ligands will help address gaps in our understanding regarding AhR-dependent toxicities. Here, we review the role of the AhR within the GI tract, novel therapeutic modality approaches to target the AhR, key AhR-dependent safety liabilities, and relevant strategies that can be implemented to address drug safety concerns. Together, this review discusses the emerging therapeutic landscape of modalities targeting the AhR for inflammatory GI indications and offers a safety roadmap for AhR drug development.
Collapse
|
28
|
Aryl hydrocarbon receptor signals in epithelial cells govern the recruitment and location of Helios + Tregs in the gut. Cell Rep 2022; 39:110773. [PMID: 35545035 DOI: 10.1016/j.celrep.2022.110773] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 12/30/2021] [Accepted: 04/12/2022] [Indexed: 01/21/2023] Open
Abstract
CD4+Foxp3+ regulatory T cells (Tregs) are essential for homeostasis in the colon, but the mechanism by which local environmental cues determine the localization of colonic Tregs is unclear. Here, we administer indigo naturalis (IN), a nontoxic phytochemical aryl hydrocarbon receptor (AhR) agonist used for treating patients with ulcerative colitis (UC) in Asia, and we show that IN increases Helios+ Tregs and MHC class II+ epithelial cells (ECs) in the colon. Interactions between Tregs and MHC class II+ ECs occur mainly near the crypt bottom in the steady state, whereas Tregs dramatically increase and shift toward the crypt top following IN treatment. Moreover, the number of CD25+ T cells is increased near the surface of ECs in IN-treated UC patients compared with that in patients treated with other therapies. We also highlight additional AhR-signaling mechanisms in intestinal ECs that determine the accumulation and localization of Helios+ Tregs in the colon.
Collapse
|
29
|
Matsuno Y, Torisu T, Umeno J, Shibata H, Hirano A, Fuyuno Y, Okamoto Y, Fujioka S, Kawasaki K, Moriyama T, Nagasue T, Zeze K, Hirakawa Y, Kawatoko S, Koga Y, Oda Y, Esaki M, Kitazono T. One-year clinical efficacy and safety of indigo naturalis for active ulcerative colitis: a real-world prospective study. Intest Res 2022; 20:260-268. [PMID: 35508956 PMCID: PMC9081999 DOI: 10.5217/ir.2021.00124] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/01/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND/AIMS Recent studies suggested a favorable effect of indigo naturalis (IN) in inducing remission for refractory ulcerative colitis (UC), however, the maintenance effect of IN for patients with UC remains unknown. Therefore, we conducted a prospective uncontrolled open-label study to analyze the efficacy and safety of IN for patients with UC. METHODS Patients with moderate to severe active UC (clinical activity index [CAI] ≥ 8) took 2 g/day of IN for 52 weeks. CAI at weeks 0, 4, 8, and 52 and Mayo endoscopic subscore (MES) and Geboes score (GS) at weeks 0, 4, and 52 were assessed. Clinical remission (CAI ≤ 4), mucosal healing (MES ≤ 1), and histological healing (GS ≤ 1) rates at each assessment were evaluated. Overall adverse events (AEs) during study period were also evaluated. The impact of IN on mucosal microbial composition was assessed using 16S ribosomal RNA gene sequences. RESULTS Thirty-three patients were enrolled. The rates of clinical remission at weeks 4, 8, and 52 were 67%, 76%, and 73%, respectively. The rates of mucosal healing at weeks 4 and 52 were 48% and 70%, respectively. AEs occurred in 17 patients (51.5%) during follow-up. Four patients (12.1%) showed severe AEs, among whom 3 manifested acute colitis. No significant alteration in the mucosal microbial composition was observed with IN treatment. CONCLUSIONS One-year treatment of moderate to severe UC with IN was effective. IN might be a promising therapeutic option for maintaining remission in UC, although the relatively high rate of AEs should be considered.
Collapse
Affiliation(s)
- Yuichi Matsuno
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takehiro Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Junji Umeno
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hiroki Shibata
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Atsushi Hirano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuta Fuyuno
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yasuharu Okamoto
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shin Fujioka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Keisuke Kawasaki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tomohiko Moriyama
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
- International Medical Department, Kyushu University Hospital, Fukuoka, Japan
| | - Tomohiro Nagasue
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Keizo Zeze
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinichiro Kawatoko
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yutaka Koga
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Motohiro Esaki
- Division of Gastroenterology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
30
|
Indigo Pulverata Levis (Chung-Dae, Persicaria tinctoria) Alleviates Atopic Dermatitis-like Inflammatory Responses In Vivo and In Vitro. Int J Mol Sci 2022; 23:ijms23010553. [PMID: 35008979 PMCID: PMC8745452 DOI: 10.3390/ijms23010553] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/31/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease associated with a type 2 T helper cell (Th2) immune response. The IndigoPulverata Levis extract (CHD) is used in traditional Southeast Asian medicine; however, its beneficial effects on AD remain uninvestigated. Therefore, we investigated the therapeutic effects of CHD in 2,4-dinitrochlorobenzene (DNCB)-induced BALB/c mice and tumor necrosis factor (TNF)-α- and interferon gamma (IFN)-γ-stimulated HaCaT cells. We evaluated immune cell infiltration, skin thickness, and the serum IgE and TNF-α levels in DNCB-induced AD mice. Moreover, we measured the expression levels of pro-inflammatory cytokines, mitogen-activated protein kinase (MAPK), and the nuclear factor-kappa B (NF-κB) in the mice dorsal skin. We also studied the effect of CHD on the translocation of NF-κB p65 and inflammatory chemokines in HaCaT cells. Our in vivo results revealed that CHD reduced the dermis and epidermis thicknesses and inhibited immune cell infiltration. Furthermore, it suppressed the proinflammatory cytokine expression and MAPK and NF-κB phosphorylations in the skin tissue and decreased serum IgE and TNF-α levels. In vitro results indicated that CHD downregulated inflammatory chemokines and blocked NF-κB p65 translocation. Thus, we deduced that CHD is a potential drug candidate for AD treatment.
Collapse
|
31
|
Kudo T, Jimbo K, Shimizu H, Iwama I, Ishige T, Mizuochi T, Arai K, Kumagai H, Uchida K, Abukawa D, Shimizu T. Qing-Dai for pediatric ulcerative colitis multicenter survey and systematic review. Pediatr Int 2022; 64:e15113. [PMID: 35831249 DOI: 10.1111/ped.15113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pediatric ulcerative colitis (UC) is more challenging to treat than adult UC. Qing-Dai therapy is effective in adults but reports of its efficacy in children are unavailable. We conducted a questionnaire survey on Qing-Dai use among pediatric patients with UC in Japan to determine its efficacy and safety. METHODS Questionnaires were sent to 31 high-volume centers treating pediatric patients with inflammatory bowel disease. The number of patients using Qing-Dai, short-term and long-term effects, and adverse events were assessed. A systematic review of studies on the efficacy and safety of Qing-Dai usage for UC was also performed. RESULTS Overall, 29/31 facilities (93.5%) responded, Qing-Dai was used in 107 patients with UC, and 84/107 patients (78.5%) initiated treatment. Within 6 months, 81/101 (80.2%) patients had clinical remission, while 59/92 (64.1%) patients had no relapse and 29/92 (31.5%) experienced only one to two relapses yearly. Eighty-seven percent of the patients underwent regular follow ups for adverse events, among whom one patient was diagnosed with pulmonary arterial hypertension (PAH), five with enteritis, and one with headache. In the systematic review, the clinical remission rate was 50-80%, and PAH was observed in 14 of 1,158 patients (1.2%). CONCLUSIONS Qing-Dai is highly effective in treating pediatric UC. However, Qing-Dai should be administered with caution as it may cause adverse events such as PAH.
Collapse
Affiliation(s)
- Takahiro Kudo
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Keisuke Jimbo
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hirotaka Shimizu
- Division of Gastroenterology, National Center for Child Health and Development, Tokyo, Japan
| | - Itaru Iwama
- Division of Gastroenterology and Hepatology, Saitama Children's Medical Center, Saitama, Japan
| | - Takashi Ishige
- Department of Pediatrics, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Tatsuki Mizuochi
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Fukuoka, Japan
| | - Katsuhiro Arai
- Division of Gastroenterology, National Center for Child Health and Development, Tokyo, Japan
| | - Hideki Kumagai
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Keiichi Uchida
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Mie, Japan
| | - Daiki Abukawa
- Department of General Pediatrics and Gastroenterology, Miyagi Children's Hospital, Miyagi, Japan
| | - Toshiaki Shimizu
- Department of Pediatrics, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Saiki JP, Andreasson JO, Grimes KV, Frumkin LR, Sanjines E, Davidson MG, Park KT, Limketkai B. Treatment-refractory ulcerative colitis responsive to indigo naturalis. BMJ Open Gastroenterol 2022; 8:bmjgast-2021-000813. [PMID: 34969665 PMCID: PMC8718466 DOI: 10.1136/bmjgast-2021-000813] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
Background Indigo naturalis (IN) is an herbal medicine that has been used for ulcerative colitis with an unclear mechanism of action. Indigo and indirubin, its main constituents, are ligands of the aryl hydrocarbon receptor (AhR). We assessed the safety, efficacy, and colon AhR activity of IN given orally to patients with treatment-refractory ulcerative colitis. The role of AhR in IN benefit was further evaluated with an AhR antagonist in a murine colitis model. Methods This open-label, dose-escalation study sequentially treated 11 patients with ulcerative colitis with either IN 500 mg/day or 1.5 g/day for 8 weeks, followed by a 4-week non-treatment period. The primary efficacy endpoint was clinical response at week 8, assessed by total Mayo score. Secondary endpoints included clinical remission, Ulcerative Colitis Endoscopic Index of Severity, quality of life, and colon AhR activity measured by cytochrome P450 1A1 (CYP1A1) RNA expression. Results Ten of 11 (91%) patients, including 8/9 (89%) with moderate-to-severe disease, achieved a clinical response. Among these 10 patients, all had failed treatment with 5-aminosalicylic acid, 8 patients with a tumour necrosis factor (TNF)-alpha inhibitor, and 6 patients with TNF-alpha inhibitor and vedolizumab. Five patients were corticosteroid dependent. Clinical response was observed in all five patients who had been recommended for colectomy. Three patients achieved clinical remission. All patients experienced improved endoscopic severity and quality of life. Four weeks after treatment completion, six patients had worsened partial Mayo scores. Four patients progressed to colectomy after study completion. Colon CYP1A1 RNA expression increased 12 557-fold at week 8 among six patients evaluated. No patient discontinued IN due to an adverse event. Concomitant administration of 3-methoxy-4-nitroflavone, an AhR antagonist, in a murine colitis model abrogated the benefit of IN. Conclusion IN is a potentially effective therapy for patients with treatment-refractory ulcerative colitis. This benefit is likely through AhR activation. Trial registration number NCT02442960.
Collapse
Affiliation(s)
- Julie P Saiki
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Johan Ol Andreasson
- Department of Genetics, Department of Biochemistry, Stanford University School of Medicine, Stanford, California, USA
| | - Kevin V Grimes
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Lyn R Frumkin
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Elvi Sanjines
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
| | | | - K T Park
- Division of Pediatric Gastroenterology, Stanford University School of Medicine, Stanford, California, USA
| | - Berkeley Limketkai
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
33
|
Kubota A, Terasaki M, Takai R, Kobayashi M, Muromoto R, Kojima H. 5-Aminosalicylic Acid, A Weak Agonist for Aryl Hydrocarbon Receptor That Induces Splenic Regulatory T Cells. Pharmacology 2021; 107:28-34. [PMID: 34915497 DOI: 10.1159/000520404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/21/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION 5-Aminosalicylic acid (5-ASA) is widely used as a key drug in inflammatory bowel disease. It has been recently reported that 5-ASA induces CD4 + Foxp3 + regulatory T cells (Tregs) in the colon via the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that regulates inflammation. However, the role of 5-ASA as an AhR agonist that induces Tregs in the spleen remains unknown. METHODS In the present study, we investigated these themes using an AhR-mediated transactivation assay and flow cytometry analysis. The experiments were conducted by using DR-EcoScreen cells and C57BL/6 mice. RESULTS The DR-EcoScreen cell-based transactivation assay revealed that 5-ASA acted as a weak AhR agonist at concentrations of ≥300 μM (1.31-1.45-fold), and that a typical AhR agonist, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), activated AhR at a concentration of 0.1 nM (22.8-fold). In addition, the treatment of mouse splenic cells with 300 μM 5-ASA in a primary culture assay significantly induced CD4+CD25 + Foxp3 + Tregs (control vs. 5-ASA: 9.0% vs. 12.65%, p < 0.05), while 0.1 nM TCDD also showed significant induction of Tregs (control vs. TCDD: 9.0% vs. 14.1%, p < 0.05). Interestingly, this induction was eliminated by co-treatment with an AhR antagonist, CH-223191. DISCUSSION These results suggest that 5-ASA is a weak agonist of AhR and thereby induces Tregs in spleen cells. Our findings may provide useful insights into the mechanism by which 5-ASA regulates inflammation.
Collapse
Affiliation(s)
- Atsuhito Kubota
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan,
| | - Masaru Terasaki
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Rie Takai
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | - Masaki Kobayashi
- Laboratory of Clinical Pharmaceutics & Therapeutics, Division of Pharma Sciences, Faculty of Pharmaceutical Sciences "Sapporo,", Hokkaido, Japan
| | - Ryuta Muromoto
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hiroyuki Kojima
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| |
Collapse
|
34
|
Shi J, Weng JH, Mitchison TJ. Immunomodulatory drug discovery from herbal medicines: Insights from organ-specific activity and xenobiotic defenses. eLife 2021; 10:e73673. [PMID: 34779403 PMCID: PMC8592567 DOI: 10.7554/elife.73673] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Traditional herbal medicines, which emphasize a holistic, patient-centric view of disease treatment, provide an exciting starting point for discovery of new immunomodulatory drugs. Progress on identification of herbal molecules with proven single agent activity has been slow, in part because of insufficient consideration of pharmacology fundamentals. Many molecules derived from medicinal plants exhibit low oral bioavailability and rapid clearance, leading to low systemic exposure. Recent research suggests that such molecules can act locally in the gut or liver to activate xenobiotic defense pathways that trigger beneficial systemic effects on the immune system. We discuss this hypothesis in the context of four plant-derived molecules with immunomodulatory activity: indigo, polysaccharides, colchicine, and ginsenosides. We end by proposing research strategies for identification of novel immunomodulatory drugs from herbal medicine sources that are informed by the possibility of local action in the gut or liver, leading to generation of systemic immune mediators.
Collapse
Affiliation(s)
- Jue Shi
- Centre for Quantitative Systems Biology, Department of Physics and Department of Biology, Hong Kong Baptist UniversityHong KongChina
| | - Jui-Hsia Weng
- Department of Systems Biology, Harvard Medical SchoolBostonUnited States
- Institute of Biological Chemistry, Academia SinicaTaipeiTaiwan
| | | |
Collapse
|
35
|
Long P, Li Y, Wen Q, Huang M, Li S, Lin Y, Huang X, Chen M, Ouyang J, Ao Y, Qi Q, Zhang H, Ye W, Cheng G, Zhang X, Zhang D. 3'-Oxo-tabernaelegantine A (OTNA) selectively relaxes pulmonary arteries by inhibiting AhR. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153751. [PMID: 34563984 DOI: 10.1016/j.phymed.2021.153751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/04/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH), characterized by pulmonary artery constriction and vascular remodeling, has a high mortality rate. New drugs for the treatment of PAH urgently need to be developed. PURPOSE This study was designed to investigate the vasorelaxant activity of OTNA in isolated pulmonary arteries, and explore its molecular mechanism. METHODS Pulmonary arteries and thoracic aortas were isolated from mice, and vascular tone was tested with a Wire Myograph System. Nitric oxide levels were determined with DAF-FM DA and DAX-J2™ Red. Cellular thermal shift assays, microscale thermophoresis, and molecular docking were used to identify the interaction between OTNA and aryl hydrocarbon receptor (AhR). The levels of PI3K, p-PI3K, Akt, p-Akt, eNOS, p-eNOS, and AhR were analyzed by Western blotting. RESULTS OTNA selectively relaxed the isolated pulmonary artery rings in an endothelium-dependent manner. Mechanistic study showed that OTNA induced NO production through activation of the PI3K/Akt/eNOS pathway in endothelial cells. Furthermore, we also found that OTNA directly bound to AhR and activated the PI3K/Akt/eNOS pathway to dilate pulmonary arteries by inhibiting AhR. CONCLUSIONS OTNA relaxes pulmonary arteries by antagonizing AhR. This study provides a new natural antagonist of AhR as a promising lead compound for PAH treatment.
Collapse
Affiliation(s)
- Pei Long
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Yong Li
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China; School of Pharmacy, North Sichuan Medical College, Nanchong 637100, China
| | - Qing Wen
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Maohua Huang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Songtao Li
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Yuning Lin
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Xiaojun Huang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Jie Ouyang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Yunlin Ao
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Qi Qi
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Haipeng Zhang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wencai Ye
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Guohua Cheng
- College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Xiaoqi Zhang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China.
| | - Dongmei Zhang
- College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
36
|
Han H, Safe S, Jayaraman A, Chapkin RS. Diet-Host-Microbiota Interactions Shape Aryl Hydrocarbon Receptor Ligand Production to Modulate Intestinal Homeostasis. Annu Rev Nutr 2021; 41:455-478. [PMID: 34633858 PMCID: PMC8667662 DOI: 10.1146/annurev-nutr-043020-090050] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated basic-helix-loop-helix transcription factor that binds structurally diverse ligands and senses cues from environmental toxicants and physiologically relevant dietary/microbiota-derived ligands. The AhR is an ancient conserved protein and is widely expressed across different tissues in vertebrates and invertebrates. AhR signaling mediates a wide range of cellular functions in a ligand-, cell type-, species-, and context-specific manner. Dysregulation of AhR signaling is linked to many developmental defects and chronic diseases. In this review, we discuss the emerging role of AhR signaling in mediating bidirectional host-microbiome interactions. We also consider evidence showing the potential for the dietary/microbial enhancement ofhealth-promoting AhR ligands to improve clinical pathway management in the context of inflammatory bowel diseases and colon tumorigenesis.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex Diseases and Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA;
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
| | - Stephen Safe
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, USA
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases and Department of Nutrition, Texas A&M University, College Station, Texas 77843, USA;
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, Texas 77843, USA
| |
Collapse
|
37
|
Bungsu I, Kifli N, Ahmad SR, Ghani H, Cunningham AC. Herbal Plants: The Role of AhR in Mediating Immunomodulation. Front Immunol 2021; 12:697663. [PMID: 34249001 PMCID: PMC8264659 DOI: 10.3389/fimmu.2021.697663] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022] Open
Abstract
The prevalence of chronic inflammatory diseases including inflammatory bowel disease (IBD), autoimmunity and cancer have increased in recent years. Herbal-based compounds such as flavonoids have been demonstrated to contribute to the modulation of these diseases although understanding their mechanism of action remains limited. Flavonoids are able to interact with cellular immune components in a distinct way and influence immune responses at a molecular level. In this mini review, we highlight recent progress in our understanding of the modulation of immune responses by the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor whose activity can be regulated by diverse molecules including flavonoids. We focus on the role of AhR in integrating signals from flavonoids to modulate inflammatory responses using in vitro and experimental animal models. We also summarize the limitations of these studies. Medicinal herbs have been widely used to treat inflammatory disorders and may offer a valuable therapeutic strategy to treat aberrant inflammatory responses by modulation of the AhR pathway.
Collapse
Affiliation(s)
- Izzah Bungsu
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Nurolaini Kifli
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Siti Rohaiza Ahmad
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Hazim Ghani
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| | - Anne Catherine Cunningham
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah (PAPRSB), Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei
| |
Collapse
|
38
|
Tsuji H, Kondo M, Odani W, Takino T, Takeda R, Sakai T. Treatment with indigo plant (Polygonum tinctorium Lour) improves serum lipid profiles in Wistar rats fed a high-fat diet. THE JOURNAL OF MEDICAL INVESTIGATION 2021; 67:158-162. [PMID: 32378600 DOI: 10.2152/jmi.67.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
We investigated the effects of Polygonum tinctorium Lour (PTL), a plant commonly known as indigo, on biological parameters in an animal model of high-fat diet-induced obesity. Wistar rats fed a high-fat diet and treated with PTL showed lower serum levels of triglycerides and total cholesterol levels and a higher serum levels of HDL cholesterol than those in Wistar rats fed a high-fat diet without PTL treatment. The weight of mesenteric fat in PTL-treated rats was decreased compared to that in control rats not treated with PTL. In addition, energy metabolic rate in the dark period, but not in the light period, in PTL-treated rats was higher than that in control rats. Although a significant difference was not observed, body weight in PTL-treated rats tended to be decreased compared to that in control rats. The results show that PTL improves serum lipid profiles in Wistar rats with high-fat diet-induced obesity. J. Med. Invest. 67 : 158-162, February, 2020.
Collapse
Affiliation(s)
- Hiroko Tsuji
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan.,Faculity of Human Life Science, Shikoku University, Oujin-cho 123-1, Tokushima 771-1192, Japan
| | - Maki Kondo
- Faculity of Human Life Science, Shikoku University, Oujin-cho 123-1, Tokushima 771-1192, Japan
| | - Wataru Odani
- Faculity of Human Life Science, Shikoku University, Oujin-cho 123-1, Tokushima 771-1192, Japan
| | - Tasuku Takino
- Faculity of Human Life Science, Shikoku University, Oujin-cho 123-1, Tokushima 771-1192, Japan
| | - Risako Takeda
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan.,Faculity of Human Life Science, Shikoku University, Oujin-cho 123-1, Tokushima 771-1192, Japan
| | - Tohru Sakai
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan
| |
Collapse
|
39
|
Sun Q, Leng J, Tang L, Wang L, Fu C. A Comprehensive Review of the Chemistry, Pharmacokinetics, Pharmacology, Clinical Applications, Adverse Events, and Quality Control of Indigo Naturalis. Front Pharmacol 2021; 12:664022. [PMID: 34135755 PMCID: PMC8200773 DOI: 10.3389/fphar.2021.664022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/10/2021] [Indexed: 01/09/2023] Open
Abstract
Indigo naturalis (IN), which is derived from indigo plants such as Strobilanthes cusia (Nees) Kuntze, Persicaria tinctoria (Aiton) Spach, and Isatis tinctoria L., has been traditionally used in the treatment of hemoptysis, epistaxis, chest pain, aphtha, and infantile convulsion in China for thousands of years. Clinical trials have shown that the curative effect of IN for psoriasis and ulcerative colitis (UC) is remarkable. A total of sixty-three compounds, including indole alkaloids, terpenoids, organic acids, steroids, and nucleosides, have been isolated from IN, of which indole alkaloids are the most important. Indirubin, isolated from IN, was used as a new agent to treat leukemia in China in the 1970s. Indirubin is also an active ingredient in the treatment of psoriasis. Pharmacological studies have confirmed that IN has inhibitory effects on inflammation, tumors, bacteria, and psoriasis. Indigo, indirubin, tryptanthrin, isorhamnetin, indigodole A, and indigodole C are responsible for these activities. This review provides up-to-date and comprehensive information on IN with regard to its chemistry, pharmacokinetics, pharmacology, clinical applications, adverse events, and quality control. This review may also serve a reference for further research on IN.
Collapse
Affiliation(s)
- Quan Sun
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Jing Leng
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Ling Tang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Lijuan Wang
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Chaomei Fu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
40
|
Shimizu T, Takagi C, Sawano T, Eijima Y, Nakatani J, Fujita T, Tanaka H. Indigo enhances wound healing activity of Caco-2 cells via activation of the aryl hydrocarbon receptor. J Nat Med 2021; 75:833-839. [PMID: 33963491 DOI: 10.1007/s11418-021-01524-y] [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: 03/17/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Indigo Naturalis, also known as Qing Dai (QD) is a compound obtained from Indigofera tinctoria, Isatis tinctoria, and Polygonum tinctoria and is known to ameliorate refractory ulcerative colitis (UC) by an unknown mechanism. QD maintains both homeostasis and the integrity of colon epithelia in mice that have experimentally induced colitis. The primary component of QD, indigo, comprises 42.4% of the compound. Indigo efficiently suppresses rectal bleeding and reduces the erosion of the colon epithelium, whereas it does not reduce weight loss or increase survival in a certain condition. Indigo is a ligand of the aryl hydrocarbon receptor (AhR), which is involved in the anti-colitis activity of QD. Here we investigate the effects of indigo on wound (erosion) closure in colon epithelial cells. Oral administration of indigo induced expression of Cytochrome P450 1A1 (Cyp1a1) in the colon but not in the liver, suggesting that indigo stimulates AhR from the luminal side of the colon. The erosion-closure activity tested in the scratch assays using Caco-2 cells was accelerated by addition of QD and indigo to the culture medium. QD and indigo also induced nuclear localization of AhR and expression of CYP1A1 in the Caco-2 cells. Acceleration of scratch wound closure was abolished by addition of the AhR-antagonist CH223191. Cell proliferation and actin polymerization were also shown to contribute to erosion closure. The results suggest that indigo exerts its erosion-healing effects by increasing proliferation and migration of colon epithelial cells via activation of AhR in intestinal epithelia.
Collapse
Affiliation(s)
- Takaaki Shimizu
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Chisa Takagi
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Toshinori Sawano
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Yuto Eijima
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Jin Nakatani
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, 525-8577, Japan
| | - Hidekazu Tanaka
- Pharmacology Laboratory, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji Higashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
41
|
Schulz-Kuhnt A, Neurath MF, Wirtz S, Atreya I. Innate Lymphoid Cells as Regulators of Epithelial Integrity: Therapeutic Implications for Inflammatory Bowel Diseases. Front Med (Lausanne) 2021; 8:656745. [PMID: 33869257 PMCID: PMC8044918 DOI: 10.3389/fmed.2021.656745] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
The occurrence of epithelial defects in the gut relevantly contributes to the pathogenesis of inflammatory bowel diseases (IBD), whereby the impairment of intestinal epithelial barrier integrity seems to represent a primary trigger as well as a disease amplifying consequence of the chronic inflammatory process. Besides epithelial cell intrinsic factors, accumulated and overwhelmingly activated immune cells and their secretome have been identified as critical modulators of the pathologically altered intestinal epithelial cell (IEC) function in IBD. In this context, over the last 10 years increasing levels of attention have been paid to the group of innate lymphoid cells (ILCs). This is in particular due to a preferential location of these rather newly described innate immune cells in close proximity to mucosal barriers, their profound capacity to secrete effector cytokines and their numerical and functional alteration under chronic inflammatory conditions. Aiming on a comprehensive and updated summary of our current understanding of the bidirectional mucosal crosstalk between ILCs and IECs, this review article will in particular focus on the potential capacity of gut infiltrating type-1, type-2, and type-3 helper ILCs (ILC1s, ILC2s, and ILC3s, respectively) to impact on the survival, differentiation, and barrier function of IECs. Based on data acquired in IBD patients or in experimental models of colitis, we will discuss whether the different ILC subgroups could serve as potential therapeutic targets for maintenance of epithelial integrity and/or mucosal healing in IBD.
Collapse
Affiliation(s)
- Anja Schulz-Kuhnt
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
42
|
Optimization of Extraction of Bioactive Compounds from Baphicacanthus cusia Leaves by Hydrophobic Deep Eutectic Solvents. Molecules 2021; 26:molecules26061729. [PMID: 33808811 PMCID: PMC8003621 DOI: 10.3390/molecules26061729] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 11/16/2022] Open
Abstract
Deep eutectic solvents (DESs) are considered as efficient and green solvents for the extraction of bioactive compounds from medicinal plants. In this work, a novel method of DES-based ultrasound-assisted extraction of bioactive compounds from Baphicacanthus cusia leaves (BCL) was established. Systematic screening and the morphology of the original and treated BCL were observed with scanning electron microscopy to determine the extraction efficiency of different solvents. The extraction conditions were optimized by Box–Behnken design (BBD) tests and the optimal extraction conditions were as follows: lactic acid/L-menthol ratio of 5: 2 (mol/mol), solid–liquid ratio of 80.0 mL/g and temperature of 60.5 °C. The extraction yields of tryptanthrin, indigo and indirubin reached 0.356, 1.744 and 0.562 mg/g, respectively. The results of a 2,2-diphenyl-1-picrylhydrazy (DPPH) radical scavenging activity test indicated the feasibility of DESs in the extraction of bioactive compounds. This study indicated that L-menthol/lactic acid was a green and efficient solvent for the extraction of bioactive compounds from BCL, and DES-based ultrasound-assisted extraction could be used as an effective application strategy for the extraction of bioactive compounds from medicinal plants.
Collapse
|
43
|
Han H, Jayaraman A, Safe S, Chapkin RS. Targeting the aryl hydrocarbon receptor in stem cells to improve the use of food as medicine. CURRENT STEM CELL REPORTS 2021; 6:109-118. [PMID: 34395177 DOI: 10.1007/s40778-020-00184-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of review Intestinal stem cells, the most rapidly proliferating adult stem cells, are exquisitely sensitive to extrinsic dietary factors. Uncontrolled regulation of intestinal stem cells is closely linked to colon tumorigenesis. This review focuses on how dietary and microbial derived cues regulate intestinal stem cell functionality and colon tumorigenesis in mouse models by targeting the aryl hydrocarbon receptor (AhR). Recent findings AhR, a ligand activated transcription factor, can integrate environmental, dietary and microbial cues to modulate intestinal stem cell proliferation, differentiation and their microenvironment, affecting colon cancer risk. Modulation of AhR activity is associated with many chronic diseases, including inflammatory bowel diseases where AhR expression is protective. Summary AhR signaling controls the maintenance and differentiation of intestinal stem cells, influences local niche factors, and plays a protective role in colon tumorigenesis. Mounting evidence suggests that extrinsic nutritional/dietary cues which modulate AhR signaling may be a promising approach to colon cancer chemoprevention.
Collapse
Affiliation(s)
- Huajun Han
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843
| | - Arul Jayaraman
- Department of Chemical Engineering, Texas A&M University, College Station, TX, 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX, 77843
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843
- Department of Nutrition, Texas A&M University, College Station, TX, 77843
| |
Collapse
|
44
|
Hu Y, Ma D, Ning S, Ye Q, Zhao X, Ding Q, Liang P, Cai G, Ma X, Qin X, Wei D. High-Quality Genome of the Medicinal Plant Strobilanthes cusia Provides Insights Into the Biosynthesis of Indole Alkaloids. FRONTIERS IN PLANT SCIENCE 2021; 12:742420. [PMID: 34659312 PMCID: PMC8515051 DOI: 10.3389/fpls.2021.742420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/26/2021] [Indexed: 05/21/2023]
Abstract
Strobilanthes cusia (Nees) Kuntze is an important plant used to process the traditional Chinese herbal medicines "Qingdai" and "Nanbanlangen". The key active ingredients are indole alkaloids (IAs) that exert antibacterial, antiviral, and antitumor pharmacological activities and serve as natural dyes. We assembled the S. cusia genome at the chromosome level through combined PacBio circular consensus sequencing (CCS) and Hi-C sequencing data. Hi-C data revealed a draft genome size of 913.74 Mb, with 904.18 Mb contigs anchored into 16 pseudo-chromosomes. Contig N50 and scaffold N50 were 35.59 and 68.44 Mb, respectively. Of the 32,974 predicted protein-coding genes, 96.52% were functionally annotated in public databases. We predicted 675.66 Mb repetitive sequences, 47.08% of sequences were long terminal repeat (LTR) retrotransposons. Moreover, 983 Strobilanthes-specific genes (SSGs) were identified for the first time, accounting for ~2.98% of all protein-coding genes. Further, 245 putative centromeric and 29 putative telomeric fragments were identified. The transcriptome analysis identified 2,975 differentially expressed genes (DEGs) enriched in phenylpropanoid, flavonoid, and triterpenoid biosynthesis. This systematic characterization of key enzyme-coding genes associated with the IA pathway and basic helix-loop-helix (bHLH) transcription factor family formed a network from the shikimate pathway to the indole alkaloid synthesis pathway in S. cusia. The high-quality S. cusia genome presented herein is an essential resource for the traditional Chinese medicine genomics studies and understanding the genetic underpinning of IA biosynthesis.
Collapse
Affiliation(s)
- Yongle Hu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Ecology and Resource Engineering, Wuyi University, Wuyishan, China
- Fujian Provincial Key Laboratory of Eco-Industrial Green Technology, Wuyishan, China
| | - Dongna Ma
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Shuju Ning
- College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qi Ye
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xuanxuan Zhao
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qiansu Ding
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Pingping Liang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, China
| | - Guoqian Cai
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaomao Ma
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xia Qin
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Daozhi Wei
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- *Correspondence: Daozhi Wei
| |
Collapse
|
45
|
Transcriptional programmes underlying cellular identity and microbial responsiveness in the intestinal epithelium. Nat Rev Gastroenterol Hepatol 2021; 18:7-23. [PMID: 33024279 PMCID: PMC7997278 DOI: 10.1038/s41575-020-00357-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 12/19/2022]
Abstract
The intestinal epithelium serves the unique and critical function of harvesting dietary nutrients, while simultaneously acting as a cellular barrier separating tissues from the luminal environment and gut microbial ecosystem. Two salient features of the intestinal epithelium enable it to perform these complex functions. First, cells within the intestinal epithelium achieve a wide range of specialized identities, including different cell types and distinct anterior-posterior patterning along the intestine. Second, intestinal epithelial cells are sensitive and responsive to the dynamic milieu of dietary nutrients, xenobiotics and microorganisms encountered in the intestinal luminal environment. These diverse identities and responsiveness of intestinal epithelial cells are achieved in part through the differential transcription of genes encoded in their shared genome. Here, we review insights from mice and other vertebrate models into the transcriptional regulatory mechanisms underlying intestinal epithelial identity and microbial responsiveness, including DNA methylation, chromatin accessibility, histone modifications and transcription factors. These studies are revealing that most transcription factors involved in intestinal epithelial identity also respond to changes in the microbiota, raising both opportunities and challenges to discern the underlying integrative transcriptional regulatory networks.
Collapse
|
46
|
Qi-Yue Y, Ting Z, Ya-Nan H, Sheng-Jie H, Xuan D, Li H, Chun-Guang X. From natural dye to herbal medicine: a systematic review of chemical constituents, pharmacological effects and clinical applications of indigo naturalis. Chin Med 2020; 15:127. [PMID: 33317592 PMCID: PMC7734464 DOI: 10.1186/s13020-020-00406-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022] Open
Abstract
Background Indigo naturalis is a blue dye in ancient, as well as an extensive used traditional Chinese medicine. It has a wide spectrum of pharmacological properties and can be used to treat numerous ailments such as leukemia, psoriasis, and ulcerative colitis. This article aims to expand our understanding of indigo naturalis in terms of its chemical constituents, pharmacological action and clinical applications. Methods We searched PubMed, web of science, CNKI, Google academic, Elsevier and other databases with the key words of “Indigo naturalis”, and reviewed and sorted out the modern research of indigo naturalis based on our research results. Results We outlined the traditional manufacturing process, chemical composition and quality control of indigo naturalis, systematically reviewed traditional applictions, pharmacological activities and mechanism of indigo naturalis, and summarized its clinical trials about treatment of psoriasis, leukemia and ulcerative colitis. Conclusions Indigo naturalis has a variety of pharmacological activities, such as anti-inflammatory, antioxidant, antibacterial, antiviral, immunomodulatory and so on. It has very good clinical effect on psoriasis, leukemia and ulcerative colitis. However, it should be noted that long-term use of indigo naturalis may produce some reversible adverse reactions. In summarize, indigo naturalis is an extremely important drug with great value and potential.![]()
Collapse
Affiliation(s)
- Yang Qi-Yue
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Chengdu, 610075, People's Republic of China
| | - Zhang Ting
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - He Ya-Nan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Huang Sheng-Jie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Deng Xuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Han Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China. .,Chengdu University of Traditional Chinese Medicine, No. 1188 Liutai Avenue, Chengdu, 611137, China.
| | - Xie Chun-Guang
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Chengdu, 610075, People's Republic of China.
| |
Collapse
|
47
|
Iwatani S, Shinzaki S, Amano T, Otake Y, Tani M, Yoshihara T, Tsujii Y, Hayashi Y, Inoue T, Okuzaki D, Mizushima T, Miyoshi E, Iijima H, Takehara T. Oligosaccharide-dependent anti-inflammatory role of galectin-1 for macrophages in ulcerative colitis. J Gastroenterol Hepatol 2020; 35:2158-2169. [PMID: 32424849 DOI: 10.1111/jgh.15097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/05/2020] [Accepted: 05/10/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIM Galectin-1 plays a protective role against colitis by binding with polylactosamine structures on macrophages in β-1,4-galactosyltransferase I-deficient mice, but the precise function of galectin-1 remains unknown. In the present study, we investigated the anti-inflammatory role of galectin-1 on macrophages to ameliorate ulcerative colitis in both animal model and human tissue samples. METHODS The expression of galectin-1 in colonic tissues of ulcerative colitis patients was evaluated by immunohistochemistry. Cytokine production of mouse bone marrow-derived macrophages (BMDMs) cultured with galectin-1 was investigated. Galectin-1 binding capacity and polylactosamine expression in macrophages stimulated with lipopolysaccharides were evaluated by flow cytometry. BMDMs cultured with galectin-1 were transferred into Recombination activating gene (Rag) 2-/- mice, and the severity of the dextran sodium sulfate-induced colitis model was investigated. Furthermore, RNA sequencing was performed to characterize macrophages treated with galectin-1. RESULTS In ulcerative colitis patients, tissue expression of galectin-1was decreased in inflamed mucosa compared with non-inflamed mucosa. Galectin-1 induced interleukin-10 production in BMDMs, and the interleukin-10 production was abrogated by lactose, which inhibits the interaction of oligosaccharide-galectin binding. Dextran sodium sulfate colitis was significantly ameliorated in Rag2-/- mice undergoing galectin-1-treated BMDM transfer compared with those undergoing vehicle-treated BMDM transfer. RNA sequencing revealed that treatment with galectin-1 increased the expression of CCAAT/enhancer binding protein β and CD163, but decreased the expression of CD80 on BMDMs. CONCLUSION Galectin-1, whose expression is decreased in the inflamed mucosa of ulcerative colitis patients, can ameliorate murine colitis by conferring oligosaccharide-dependent anti-inflammatory properties to macrophages.
Collapse
Affiliation(s)
- Shuko Iwatani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahiro Amano
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuriko Otake
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Mizuki Tani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takeo Yoshihara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshiki Tsujii
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshito Hayashi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takahiro Inoue
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hideki Iijima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
48
|
Song D, Lai L, Ran Z. Metabolic Regulation of Group 3 Innate Lymphoid Cells and Their Role in Inflammatory Bowel Disease. Front Immunol 2020; 11:580467. [PMID: 33193381 PMCID: PMC7649203 DOI: 10.3389/fimmu.2020.580467] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic and relapsing inflammatory disorder of the intestine. IBD is associated with complex pathogenesis, and considerable data suggest that innate lymphoid cells contribute to the development and progression of the condition. Group 3 innate lymphoid cells (ILC3s) not only play a protective role in maintaining intestinal homeostasis and gut barrier function, but also a pathogenic role in intestinal inflammation. ILC3s can sense environmental and host-derived signals and combine these cues to modulate cell expansion, migration and function, and transmit information to the broader immune system. Herein, we review current knowledge of how ILC3s can be regulated by dietary nutrients, microbiota and their metabolites, as well as other metabolites. In addition, we describe the phenotypic and functional alterations of ILC3s in IBD and discuss the therapeutic potential of ILC3s in the treatment of IBD.
Collapse
Affiliation(s)
| | | | - Zhihua Ran
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
49
|
Kimura H, Tokuyama-Nakai S, Hirabayashi Y, Ishihara T, Jisaka M, Yokota K. Anti-inflammatory and bioavailability studies on dietary 3,5,4'-trihydroxy-6,7-methylenedioxyflavone-O-glycosides and their aglycone from indigo leaves in a murine model of inflammatory bowel disease. J Pharm Biomed Anal 2020; 193:113716. [PMID: 33152603 DOI: 10.1016/j.jpba.2020.113716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 01/21/2023]
Abstract
Persicaria tinctoria (Aiton) Spach, also called Polygonum tinctorium Lour., (family Polygonaceae) for indigo plant has been traditionally useful as a medicinal or edible plant with a variety of biological activities. Of these, much attention has been paid to their anti-inflammatory activities. We have recently demonstrated that indigo leaves contain high levels of flavonol O-glycosides with 3,5,4'-trihydroxy-6,7-methylenedioxyflavone (TMF) as an aglycone. In this study, we attempted to evaluate anti-inflammatory activities of TMF-O-glycosides and free TMF prepared from indigo leaves after extraction with hot water. Free TMF was found to appreciably down-regulate the gene expression of pro-inflammatory cytokines including interleukin (IL)-1β, IL-6, inducible nitric oxide, and tumor necrosis factor-α in cultured macrophage cells stimulated with lipopolysaccharide while up-regulating the expression of anti-inflammatory IL-10. However, no study has been conducted regarding in vivo anti-inflammatory activities of TMF-O-glycosides and free TMF until now. Here, we assessed in vivo anti-inflammatory effects of these dietary compounds on ulcerative colitis in a murine model of inflammatory bowel disease by the induction with dextran sulfate sodium (DSS). Histological evaluation revealed that both TMF-O-glycosides and free TMF effectively protected against DSS-induced ulcerative colitis. The analysis of digested products by liquid chromatography and mass spectrometry led us to detect free TMF as a predominant metabolite in the feces of mice fed with TMF-O-glycosides. Moreover, free TMF was later detected as glucuronyl conjugates of TMF in the liver of mice fed with both fractions. These results indicate the effective digestion of TMF-O-glycosides and the subsequent absorption of free TMF in the gut of mice for exerting anti-inflammatory effects. Taken together, our findings suggest that dietary TMF-O-glycosides could be promising natural sources for the utilization as herbal medicine and nutraceuticals to expect in vivo anti-inflammatory activities.
Collapse
Affiliation(s)
- Hideto Kimura
- Department of Research and Development, Kotobuki Seika Co., Ltd., 2028 Hatagasaki, Yonago, Tottori 683-0845, Japan
| | - Shota Tokuyama-Nakai
- Department of Research and Development, Kotobuki Seika Co., Ltd., 2028 Hatagasaki, Yonago, Tottori 683-0845, Japan
| | - Yu Hirabayashi
- Department of Research and Development, Kotobuki Seika Co., Ltd., 2028 Hatagasaki, Yonago, Tottori 683-0845, Japan
| | - Tomoe Ishihara
- Department of Research and Development, Kotobuki Seika Co., Ltd., 2028 Hatagasaki, Yonago, Tottori 683-0845, Japan
| | - Mitsuo Jisaka
- Department of Life Science and Biotechnology, Faculty of Life and Environmental Science, Shimane University, 1060 Nishikawatsu-cho, Matsue, Shimane 690-8504, Japan; The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Kazushige Yokota
- Department of Life Science and Biotechnology, Faculty of Life and Environmental Science, Shimane University, 1060 Nishikawatsu-cho, Matsue, Shimane 690-8504, Japan; The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan.
| |
Collapse
|
50
|
Dvorak Z, Klapholz M, Burris TP, Willing BP, Gioiello A, Pellicciari R, Galli F, March J, O'Keefe SJ, Sartor RB, Kim CH, Levy M, Mani S. Weak Microbial Metabolites: a Treasure Trove for Using Biomimicry to Discover and Optimize Drugs. Mol Pharmacol 2020; 98:343-349. [PMID: 32764096 PMCID: PMC7485585 DOI: 10.1124/molpharm.120.000035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
For decades, traditional drug discovery has used natural product and synthetic chemistry approaches to generate libraries of compounds, with some ending as promising drug candidates. A complementary approach has been to adopt the concept of biomimicry of natural products and metabolites so as to improve multiple drug-like features of the parent molecule. In this effort, promiscuous and weak interactions between ligands and receptors are often ignored in a drug discovery process. In this Emerging Concepts article, we highlight microbial metabolite mimicry, whereby parent metabolites have weak interactions with their receptors that then have led to discrete examples of more potent and effective drug-like molecules. We show specific examples of parent-metabolite mimics with potent effects in vitro and in vivo. Furthermore, we show examples of emerging microbial ligand-receptor interactions and provide a context in which these ligands could be improved as potential drugs. A balanced conceptual advance is provided in which we also acknowledge potential pitfalls-hyperstimulation of finely balanced receptor-ligand interactions could also be detrimental. However, with balance, we provide examples of where this emerging concept needs to be tested. SIGNIFICANCE STATEMENT: Microbial metabolite mimicry is a novel way to expand on the chemical repertoire of future drugs. The emerging concept is now explained using specific examples of the discovery of therapeutic leads from microbial metabolites.
Collapse
Affiliation(s)
- Zdenek Dvorak
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Max Klapholz
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Thomas P Burris
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Benjamin P Willing
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Antimo Gioiello
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Roberto Pellicciari
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Francesco Galli
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - John March
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Stephen J O'Keefe
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - R Balfour Sartor
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Chang H Kim
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Maayan Levy
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| | - Sridhar Mani
- Department of Cell Biology and Genetics, Palacký University, Olomouc, Czech Republic (Z.D.); Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania (M.K., M.L.); The Center for Clinical Pharmacology, Washington University in St. Louis and St. Louis College of Pharmacy, St. Louis, Missouri (T.P.B.); Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta (B.P.W.); Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy (A.G., F.G.); TES Pharma, Corso Vannucci, Perugia, Italy (R.P.); The Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York (J.M.); Division of Gastroenterology and Nutrition, UPMC Presbyterian Hospital, Pittsburgh, Pennsylvania (S.J.O.); Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.B.S.); Department of Pathology, Mary H. Weiser Food Allergy Center, and Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan (C.H.K.); and Department of Medicine, Albert Einstein College of Medicine, Bronx, New York (S.M.)
| |
Collapse
|