1
|
Gupta J, Almulla AF, Jalil AT, Jasim NY, Aminov Z, Alsaikhan F, Ramaiah P, Chinnasamy L, Jawhar ZH. Melatonin in Chemo/Radiation Therapy; Implications for Normal Tissues Sparing and Tumor Suppression: An Updated Review. Curr Med Chem 2025; 32:511-538. [PMID: 37916636 DOI: 10.2174/0109298673262122231011172100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/20/2023] [Accepted: 09/01/2023] [Indexed: 11/03/2023]
Abstract
Resistance to therapy and the toxicity of normal tissue are the major problems for efficacy associated with chemotherapy and radiotherapy. Drug resistance is responsible for most cases of mortality associated with cancer. Furthermore, their side effects can decrease the quality of life for surviving patients. An enhancement in the tumor response to therapy and alleviation of toxic effects remain unsolved challenges. One of the interesting topics is the administration of agents with low toxicity to protect normal tissues and/or sensitize cancers to chemo/radiotherapy. Melatonin is a natural body hormone that is known as a multitasking molecule. Although it has antioxidant properties, a large number of experiments have uncovered interesting effects of melatonin that can increase the therapeutic efficacy of chemo/radiation therapy. Melatonin can enhance anticancer therapy efficacy through various mechanisms, cells such as the immune system, and modulation of cell cycle and death pathways, tumor suppressor genes, and also through suppression of some drug resistance mediators. However, melatonin may protect normal tissues through the suppression of inflammation, fibrosis, and massive oxidative stress in normal cells and tissues. In this review, we will discuss the distinct effects of melatonin on both tumors and normal tissues. We review how melatonin may enhance radio/chemosensitivity of tumors while protecting normal tissues such as the lung, heart, gastrointestinal system, reproductive system, brain, liver, and kidney.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | | | | | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
2
|
Liang W, Ren Y, Wang Y, Chen W, Mo Z, Yang C, Nie K. Xiao-Ban-Xia Decoction Alleviates Chemotherapy-Induced Nausea and Vomiting by Inhibiting Ferroptosis via Activation of The Nrf2/SLC7A11/GPX4 Pathway. Adv Biol (Weinh) 2024; 8:e2400323. [PMID: 39501722 DOI: 10.1002/adbi.202400323] [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: 06/11/2024] [Revised: 10/17/2024] [Indexed: 12/14/2024]
Abstract
Chemotherapy-induced nausea and vomiting (CINV) represents the common gastrointestinal side effect for cancer patients. Xiao-Ban-Xia decoction (XBXD), a classical anti-emetic traditional Chinese medicine formula, is frequently used for the clinical treatment of CINV. This study used a cisplatin-induced rat pica model to explore whether the anti-emetic mechanism of XBXD in treating CINV is related to ferroptosis. The inflammatory damage of the gastrointestinal tract is evaluated by HE staining and ELISA. The degree of ferroptosis are validated by the iron deposition, the levels of ROS, MDA, and GSH, and the ultrastructure of mitochondria in the gastric antrum and ileum. The potential ferroptosis-related targets of XBXD against CINV are screened by network pharmacology and further assessed by Western blot. XBXD significantly decreased the kaolin consumption in rats, and improved the inflammatory pathological damage, with decreased levels of HMGB1, IL-1β, and TNF-α. Furthermore, XBXD significantly suppressed ferroptosis, as indicated by the improvement of iron deposition, mitochondrial abnormalities, and oxidative stress. The network pharmacology and Western blot results indicated that XBXD activated the Nrf2/SLC7A11/GPX4 signaling pathway. This study proved that XBXD activates the Nrf2/SLC7A11/GPX4 signaling pathway, thereby inhibiting ferroptosis, which represents a critical anti-emetic mechanism of XBXD in combatting CINV.
Collapse
Affiliation(s)
- Wan Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Yuke Ren
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Yusu Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Weijian Chen
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Ziyao Mo
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Chenglu Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| | - Ke Nie
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, P. R. China
| |
Collapse
|
3
|
Zhang Y, Wang H, Sang Y, Liu M, Wang Q, Yang H, Li X. Gut microbiota in health and disease: advances and future prospects. MedComm (Beijing) 2024; 5:e70012. [PMID: 39568773 PMCID: PMC11577303 DOI: 10.1002/mco2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
The gut microbiota plays a critical role in maintaining human health, influencing a wide range of physiological processes, including immune regulation, metabolism, and neurological function. Recent studies have shown that imbalances in gut microbiota composition can contribute to the onset and progression of various diseases, such as metabolic disorders (e.g., obesity and diabetes) and neurodegenerative conditions (e.g., Alzheimer's and Parkinson's). These conditions are often accompanied by chronic inflammation and dysregulated immune responses, which are closely linked to specific forms of cell death, including pyroptosis and ferroptosis. Pathogenic bacteria in the gut can trigger these cell death pathways through toxin release, while probiotics have been found to mitigate these effects by modulating immune responses. Despite these insights, the precise mechanisms through which the gut microbiota influences these diseases remain insufficiently understood. This review consolidates recent findings on the impact of gut microbiota in these immune-mediated and inflammation-associated conditions. It also identifies gaps in current research and explores the potential of advanced technologies, such as organ-on-chip models and the microbiome-gut-organ axis, for deepening our understanding. Emerging tools, including single-bacterium omics and spatial metabolomics, are discussed for their promise in elucidating the microbiota's role in disease development.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Hong Wang
- School of Traditional Chinese Medicine Southern Medical University Guangzhou China
| | - Yiwei Sang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Mei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Qing Wang
- School of Life Sciences Beijing University of Chinese Medicine Beijing China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs China Academy of Chinese Medical Sciences Beijing China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| |
Collapse
|
4
|
Ng CYJ, Zhong L, Ng HS, Goh KS, Zhao Y. Managing Type 2 Diabetes Mellitus via the Regulation of Gut Microbiota: A Chinese Medicine Perspective. Nutrients 2024; 16:3935. [PMID: 39599721 PMCID: PMC11597546 DOI: 10.3390/nu16223935] [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: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder characterized by insulin resistance and inadequate insulin production. Given the increased frequency of T2DM and the health issues it can cause, there is an increasing need to develop alternative T2DM management strategies. One such approach is Chinese Medicine (CM), a complementary therapy widely used in T2DM treatment. Given the emphasis on gut microbiota in current research, studying CM in the treatment of T2DM via gut microbiota modulation could be beneficial. Scope and approach: The use of various CM methods for managing T2DM via gut microbiota modulation is highlighted in this review. Following an introduction of the gut microbiota and its role in T2DM pathogenesis, we will review the potential interactions between gut microbiota and T2DM. Thereafter, we will review various CM treatment modalities that modulate gut microbiota and provide perspectives for future research. Key findings and discussion: In T2DM, Akkermansia, Bifidobacterium, and Firmicutes are examples of gut microbiota commonly imbalanced. Studies have shown that CM therapies can modulate gut microbiota, leading to beneficial effects such as reduced inflammation, improved metabolism, and improved immunity. Among these treatment modalities, Chinese Herbal Medicine and acupuncture are the most well-studied, and several in vivo studies have demonstrated their potential in managing T2DM by modulating gut microbiota. However, the underlying biomolecular mechanisms of actions are not well elucidated, which is a key area for future research. Future studies could also investigate alternate CM therapies such as moxibustion and CM exercises and conduct large-scale clinical trials to validate their effectiveness in treatment.
Collapse
Affiliation(s)
- Chester Yan Jie Ng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Linda Zhong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Han Seong Ng
- Singapore General Hospital, Outram Rd., Singapore 169608, Singapore
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
| | - Kia Seng Goh
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
- Singapore College of Traditional Chinese Medicine, 640 Lor 4 Toa Payoh, Singapore 319522, Singapore
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
| |
Collapse
|
5
|
Bae SJ, Jang Y, Kim Y, Park JH, Jang JH, Oh JY, Jang SY, Ahn S, Park HJ. Gut Microbiota Regulation by Acupuncture and Moxibustion: A Systematic Review and Meta-Analysis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1245-1273. [PMID: 39192678 DOI: 10.1142/s0192415x24500502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
There have been numerous studies investigating the impact of acupuncture and/or moxibustion on the gut microbiota, but the results have been inconclusive. Therefore, we conducted a systematic review and meta-analysis that included both preclinical and clinical studies to assess the current evidence regarding the effects of acupuncture on gut microbiota changes. We collected relevant studies from EMBASE and PubMed, collected outcomes including diversity and relative abundance measures of the gut microbiome, and the summarized effect estimates were calculated using the ratio of means (ROM) with 95% confidence intervals. Our analysis identified three clinical studies and 20 preclinical studies, encompassing various diseases and models, including colitis and obesity. The pooled results indicated no significant difference in alpha diversity changes between treatment groups and controls, except for the Simpson index measure, which was significantly higher in the treatment groups. Additionally, the pooled results showed an increase in the Firmicutes and a decrease in the Bacteroidetes in the treatment groups, along with increases in the Lactobacillus and Ruminococcus genera. These findings suggest acupuncture treatment can target the modification of specific phyla and genera of gut microbiota. However, it is important to note that the effects of acupuncture on the gut microbiome are heterogeneous across studies, particularly in different disease models.
Collapse
Affiliation(s)
- Sun-Jeong Bae
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yumi Jang
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Yejin Kim
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Ji-Han Park
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Jae-Hwan Jang
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, 128 Beobwon-ro, Songpa-gu, Seoul 05854, Republic of Korea
| | - Ju-Young Oh
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sun-Young Jang
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sora Ahn
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hi-Joon Park
- Acupuncture and Meridian Science Research Center (AMSRC), Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Korean Medical Science, Graduate School of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of KHU-KIST Convergence Science & Technology, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 027932, Republic of Korea
| |
Collapse
|
6
|
Yin Y, Li H, Qin Y, Chen T, Zhang Z, Lu G, Shen J, Shen M. Moxibustion mitigates mitochondrial dysfunction and NLRP3 inflammatory activation in cyclophosphamide-induced premature ovarian insufficiency rats. Life Sci 2023; 314:121283. [PMID: 36528078 DOI: 10.1016/j.lfs.2022.121283] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
AIMS This study aimed to investigate the protective effects of moxibustion on ovarian dysfunction in rats with cyclophosphamide (Cy)-induced premature ovarian insufficiency (POI). It also aimed at revealing its potential mechanisms and emphasizing its role in mitigating the mitochondrial dysfunction and NLRP3 inflammatory activation. MATERIALS AND METHODS POI models were established by the intraperitoneal administration of Cy using female Sprague-Dawley rats. Moxibustion (BL23 or CV4, CV8) was used to treat POI models for fifteen days. Vaginal smears, enzyme-linked immunosorbent assay, hematoxylin-eosin, tunnel staining, flow cytometry analysis, immunohistochemistry staining, qRT-PCR, and western blotting were conducted to evaluate the ovarian function, mitochondrial dysfunction, and NLRP3 inflammatory activation in this study. KEY FINDINGS Moxibustion could improve the disorder of the estrous cycles and reproductive hormone levels, promote follicular growth, reduce the number of atresia follicles, and alleviate the apoptosis of ovarian granulosa cells (GCs) in rats with POI. Furthermore, moxibustion mitigated the mitochondrial damage, reversed the elevated serum levels of IL-18 and IL-1β, and decreased their protein expression in the ovaries of rats with POI. Moxibustion significantly inhibited the expression of the mRNAs and proteins of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), caspase 1, and gasdermin D (GSDMD) in the ovaries of rats with POI. SIGNIFICANCE These results supported that moxibustion may ameliorate Cy-induced POI by mitigating the mitochondrial dysfunction and NLRP3 inflammatory activation. Targeted treatment of mitochondrial damage and NLRP3 inflammatory activation may be a novel therapeutic strategy for POI.
Collapse
Affiliation(s)
- Yaoli Yin
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongxiao Li
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yantong Qin
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ting Chen
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhizi Zhang
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ge Lu
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Shen
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Meihong Shen
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, China.
| |
Collapse
|
7
|
Zheng X, Mai L, Xu Y, Wu M, Chen L, Chen B, Su Z, Chen J, Chen H, Lai Z, Xie Y. Brucea javanica oil alleviates intestinal mucosal injury induced by chemotherapeutic agent 5-fluorouracil in mice. Front Pharmacol 2023; 14:1136076. [PMID: 36895947 PMCID: PMC9990700 DOI: 10.3389/fphar.2023.1136076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Background: Brucea javanica (L.) Merr, has a long history to be an anti-dysentery medicine for thousand of years, which is commonly called "Ya-Dan-Zi" in Chinese. The common liquid preparation of its seed, B. javanica oil (BJO) exerts anti-inflammatory action in gastrointestinal diseases and is popularly used as an antitumor adjuvant in Asia. However, there is no report that BJO has the potential to treat 5-Fluorouracil (5-FU)-induced chemotherapeutic intestinal mucosal injury (CIM). Aim of the study: To test the hypothesis that BJO has potential intestinal protection on intestinal mucosal injury caused by 5-FU in mice and to explore the mechanisms. Materials and methods: Kunming mice (half male and female), were randomly divided into six groups: normal group, 5-FU group (5-FU, 60 mg/kg), LO group (loperamide, 4.0 mg/kg), BJO group (0.125, 0.25, 0.50 g/kg). CIM was induced by intraperitoneal injection of 5-FU at a dose of 60 mg/kg/day for 5 days (from day 1 to day 5). BJO and LO were given orally 30 min prior to 5-FU administration for 7 days (from day 1 to day 7). The ameliorative effects of BJO were assessed by body weight, diarrhea assessment, and H&E staining of the intestine. Furthermore, the changes in oxidative stress level, inflammatory level, intestinal epithelial cell apoptosis, and proliferation, as well as the amount of intestinal tight junction proteins were evaluated. Finally, the involvements of the Nrf2/HO-1 pathway were tested by western blot. Results: BJO effectively alleviated 5-FU-induced CIM, as represented by the improvement of body weight, diarrhea syndrome, and histopathological changes in the ileum. BJO not only attenuated oxidative stress by upregulating SOD and downregulating MDA in the serum, but also reduced the intestinal level of COX-2 and inflammatory cytokines, and repressed CXCL1/2 and NLRP3 inflammasome activation. Moreover, BJO ameliorated 5-FU-induced epithelial apoptosis as evidenced by the downregulation of Bax and caspase-3 and the upregulation of Bcl-2, but enhanced mucosal epithelial cell proliferation as implied by the increase of crypt-localized proliferating cell nuclear antigen (PCNA) level. Furthermore, BJO contributed to the mucosal barrier by raising the level of tight junction proteins (ZO-1, occludin, and claudin-1). Mechanistically, these anti-intestinal mucositis pharmacological effects of BJO were relevant for the activation of Nrf2/HO-1 in the intestinal tissues. Conclusion: The present study provides new insights into the protective effects of BJO against CIM and suggests that BJO deserves to be applied as a potential therapeutic agent for the prevention of CIM.
Collapse
Affiliation(s)
- Xinghan Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, Guangdong, China.,Pharmacy Department, Quanzhou Hospital of Traditional Chinese Medicine, Quanzhou, China
| | - Liting Mai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Medical Insurance Office, Zhaoqing Hospital, Sun Yat-sen University, Zhaoqing, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Ying Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Minghui Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Li Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Baoyi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Hongying Chen
- Guangzhou Baiyunshan Mingxing Pharmaceutical Co. Ltd, Guangzhou, China
| | - Zhengquan Lai
- Department of Pharmacy, Shenzhen University General Hospital/Shenzhen University Clinical Medical Academy, Shenzhen University, Shenzhen, Guangdong, China
| | - Youliang Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| |
Collapse
|
8
|
Huang J, Hwang AYM, Jia Y, Kim B, Iskandar M, Mohammed AI, Cirillo N. Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models. Int J Mol Sci 2022; 23:15434. [PMID: 36499758 PMCID: PMC9737148 DOI: 10.3390/ijms232315434] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a common and most debilitating complication associated with the cytotoxicity of chemotherapy. The condition affects the entire alimentary canal from the mouth to the anus and has a significant clinical and economic impact. Although oral and intestinal mucositis can occur concurrently in the same individual, these conditions are often studied independently using organ-specific models that do not mimic human disease. Hence, the purpose of this scoping review was to provide a comprehensive yet systematic overview of the animal models that are utilised in the study of chemotherapy-induced mucositis. A search of PubMed/MEDLINE and Scopus databases was conducted to identify all relevant studies. Multiple phases of filtering were conducted, including deduplication, title/abstract screening, full-text screening, and data extraction. Studies were reported according to the updated Preferred Reporting Items for Systematic reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) guidelines. An inter-rater reliability test was conducted using Cohen's Kappa score. After title, abstract, and full-text screening, 251 articles met the inclusion criteria. Seven articles investigated both chemotherapy-induced intestinal and oral mucositis, 198 articles investigated chemotherapy-induced intestinal mucositis, and 46 studies investigated chemotherapy-induced oral mucositis. Among a total of 205 articles on chemotherapy-induced intestinal mucositis, 103 utilised 5-fluorouracil, 34 irinotecan, 16 platinum-based drugs, 33 methotrexate, and 32 other chemotherapeutic agents. Thirteen articles reported the use of a combination of 5-fluorouracil, irinotecan, platinum-based drugs, or methotrexate to induce intestinal mucositis. Among a total of 53 articles on chemotherapy-induced oral mucositis, 50 utilised 5-fluorouracil, 2 irinotecan, 2 methotrexate, 1 topotecan and 1 with other chemotherapeutic drugs. Three articles used a combination of these drugs to induce oral mucositis. Various animal models such as mice, rats, hamsters, piglets, rabbits, and zebrafish were used. The chemotherapeutic agents were introduced at various dosages via three routes of administration. Animals were mainly mice and rats. Unlike intestinal mucositis, most oral mucositis models combined mechanical or chemical irritation with chemotherapy. In conclusion, this extensive assessment of the literature revealed that there was a large variation among studies that reproduce oral and intestinal mucositis in animals. To assist with the design of a suitable preclinical model of chemotherapy-induced alimentary tract mucositis, animal types, routes of administration, dosages, and types of drugs were reported in this study. Further research is required to define an optimal protocol that improves the translatability of findings to humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, Carlton, VIC 3053, Australia
| |
Collapse
|
9
|
Bullard BM, McDonald SJ, Cardaci TD, VanderVeen BN, Murphy EA. Nonpharmacological approaches for improving gut resilience to chemotherapy. Curr Opin Support Palliat Care 2022; 16:151-160. [PMID: 35862879 DOI: 10.1097/spc.0000000000000599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Mucositis of the gastrointestinal tract is a debilitating side effect of chemotherapy that negatively influences treatment tolerance and patient life quality. This review will evaluate the recent literature on nonpharmacological strategies that have the potential to improve chemotherapy-induced mucositis (CIM). RECENT FINDINGS Alternatives to pharmacological approaches have shown great promise in preventing CIM. Natural products, including curcumin, ginseng, quercetin, and patchouli all show potential in mitigating CIM. In addition, dietary patterns, such as the elemental diet, high fiber diet, and diets high in amino acids have documented benefits in preventing CIM. Perhaps the greatest advancement coming to this arena in recent years is in the field of probiotics. Indeed, research on single species as well as probiotic mixtures show potential in reducing CIM insofar as probiotics are now being suggested for treatment of CIM by governing bodies. Although behavioral interventions including psychological interventions and exercise interventions have shown promise in reducing cancer therapy-related side effects, more work in this domain is warranted and particularly in the context of CIM. SUMMARY Alternatives to pharmacological approaches show great potential for use in prevention and treatment of CIM and should be further developed for use in the clinic.
Collapse
Affiliation(s)
- Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina, USA
| | | | | | | | | |
Collapse
|
10
|
Chen KJ, Huang YL, Kuo LM, Chen YT, Hung CF, Hsieh PW. Protective role of casuarinin from Melastoma malabathricum against a mouse model of 5-fluorouracil-induced intestinal mucositis: Impact on inflammation and gut microbiota dysbiosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154092. [PMID: 35430483 DOI: 10.1016/j.phymed.2022.154092] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND 5-FU-induced intestinal mucositis (FUIIM) is a common gastrointestinal side effect of chemotherapy, leading to gastric pain in clinical cancer patients. In a previous study, we demonstrated that neutrophil elastase (NE) inhibitors could alleviate FUIIM and manipulate the homeostasis of the gut microbiota. The root of Melastoma malabathricum, also called Ye-Mu-Dan, has been used as a traditional Chinese medicine for gastrointestinal disease. Water extract of the roots of M. malabathricum exhibits an inhibitory effect on NE, with an IC50 value of 9.13 μg/ml. PURPOSE In this study, we aimed to isolate an anti-NE compound from the root of M. malabathricum and to determine the protective effect of the bioactive component on a mouse model of FUIIM with respect to tissue damage, inflammation, intestinal barrier dysfunction, and gut microbiota dysbiosis. METHODS A water extract of the roots of M. malabathricum was prepared and its major bioactive compound, was identified using bioactivity-guided fractionation. The effects of samples on the inhibition of NE activity were evaluated using enzymatic assays. To evaluate the effects of the bioactive compound in an FUIIM animal model, male C57BL/6 mice treated with or without casuarinin (50 and 100 mg/kg/day, p.o.), and then received of 5-fluorouracil (50 mg/kg/day) intraperitoneally for 5 days to induce FUIIM. Histopathological staining was used to monitor the tissue damage, proliferation of intestinal crypts, and expression of tight junction proteins. The inflammation score was estimated by determining the levels of oxidative stress, neutrophil-related proteases, and proinflammatory cytokines in tissue and serum. The ecology of the gut microbiota was evaluated using 16S rRNA gene sequencing. RESULTS Casuarinin had the most potent and selective effect against NE, with an IC50 value of 2.79 ± 0.07 μM. Casuarinin (100 mg/kg/day, p.o.) significantly improved 5-FU-induced body weight loss together with food intake reduction, and it also significantly reversed villus atrophy, restored the proliferative activity of the intestinal crypts, and suppressed inflammation and intestinal barrier dysfunction in the mouse model of FUIIM. Casuarinin also reversed 5-FU-induced gut microbiota dysbiosis, particularly the abundance of Actinobacteria, Candidatus Arthromitus, and Lactobacillus murinus, and the Firmicutes-to-Bacteroidetes ratio. CONCLUSION This study firstly showed that casuarinin isolated from the root part of M. malabathricum could be used as a NE inhibitor, whereas it could improve FUIIM by modulating inflammation, intestinal barrier dysfunction, and gut microbiota dysbiosis. In summary, exploring anti-NE natural product may provide a way to find candidate for improvement of FUIIM.
Collapse
Affiliation(s)
- Kung-Ju Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan
| | - Yu-Ling Huang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan
| | - Liang-Mou Kuo
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yi-Ting Chen
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Program in Pharmaceutical Biotechnology, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Pei-Wen Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan 333, Taiwan; Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
11
|
Yao J, Yan X, Chen L, Li Y, Zhang L, Chen M, Li Y. Efficacy and MicroRNA-Gut Microbiota Regulatory Mechanisms of Acupuncture for Severe Chronic Constipation: Study Protocol for a Randomized Controlled Trial. Front Med (Lausanne) 2022; 9:906403. [PMID: 35836948 PMCID: PMC9273765 DOI: 10.3389/fmed.2022.906403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundSevere chronic constipation (SCC) is a common functional gastrointestinal (GI) disorder associated with disruptions in GI motility. Abnormalities between gut microbiota and microRNAs (miRNAs) are implicated in the pathogenesis of GI motility in SCC. Acupuncture has been shown to improve constipation-related symptoms and rebalance the gut microbiota. This protocol proposed a plan to explore the hypothesis that the efficacy of acupuncture is associated with the crosstalk between gut microbes and miRNAs in patients with SCC.MethodsThis trial is designed as a randomized, sham-controlled trial involving 80 patients and 40 healthy volunteers. A total of 80 patients with SCC (≤2 mean spontaneous, complete bowel movements per week [CSBMs]) will be randomly allocated to receive either 16-session acupuncture at true acupoints or non-penetrating sham acupuncture at non-acupoints for 4 weeks. The primary outcome will be the proportion of patients with ≥3 mean weekly CSBMs over weeks 1–4 and 5–8. Secondary efficacy endpoints include bowel movements, stool consistency, degree of straining, and the quality of life. Healthy volunteers will not receive any clinical intervention. Fasting plasma and fecal samples will be analyzed by 16S rRNA third-generation sequencing and miRNA high-throughput sequencing technologies. Finally, a tripartite network analysis will be used to investigate the interactions among clinical efficacy, miRNAs, and intestinal microbiota.DiscussionFrom the perspective of microRNA-gut microbiota regulatory mechanisms, our results will partially illuminate the crucial role of fecal miRNAs and intestinal microbiota to understand how acupuncture exerts its anti-constipation role.Trial registrationThis trial is registered with ChiCTR2100048831, registered 18 July 2021; ethical approval has been obtained from the Sichuan Regional Ethics Review of Committee on Traditional Chinese Medicine, approval ID: 2021KL-023.
Collapse
Affiliation(s)
- Junpeng Yao
- Acupuncture and Tuina School/ the 3 Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangyun Yan
- Acupuncture and Tuina School/ the 3 Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liping Chen
- Acupuncture and Tuina School/ the 3 Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanqiu Li
- Acupuncture and Tuina School/ the 3 Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Leixiao Zhang
- Department of Integrated Traditional and Western Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Min Chen
- Clinical Medicine School, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Li
- Acupuncture and Tuina School/ the 3 Teaching Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Ying Li
| |
Collapse
|
12
|
Liu Y, Du X, Zhai S, Tang X, Liu C, Li W. Gut microbiota and atopic dermatitis in children: a scoping review. BMC Pediatr 2022; 22:323. [PMID: 35655175 PMCID: PMC9161518 DOI: 10.1186/s12887-022-03390-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/05/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gut microbiota plays an important role in the development of atopic dermatitis (AD). We aimed to elucidate research trends in gut microbiota and AD in children, to provide evidence and insights to the clinical prevention and treatment of AD in children. METHODS A scoping literature review on the studies of gut microbiota and AD were conducted. Two authors independently searched Pubmed et al. databases for studies focused on gut microbiota and AD in children up to January 15, 2022. The literatures were screened and analyzed by two reviewers. RESULTS A total of 44 reports were finally included and analyzed. Current researches have indicated that abnormal human microecology is closely associated with AD, and the disturbance of intestinal microbiota plays an important role in the occurrence and development of AD. Probiotics can correct the microbiota disorder, have the functions of regulating immunity, antioxidant, and help to restore the microecological homeostasis. However, there is still a lack of high-quality research reports on the efficacy and safety of probiotics in the prevention and treatment of AD in children. CONCLUSIONS The changes of gut microbiota are essential to the development of AD in children, which may be an effective target for the prevention and treatment of AD. Future studies with larger sample size and rigorous design are needed to elucidate the effects and safety of probiotics in AD.
Collapse
Affiliation(s)
- Yue Liu
- Department of Acupuncture and Tuina Science, School of Traditional Chinese Medicine, Naval Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Xiaofan Du
- Clinical Medicine Science, Anhui Medical University, Hefei, 230032, China
| | - Shujie Zhai
- Department of Acupuncture and Tuina Science, School of Traditional Chinese Medicine, Naval Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Xiaodong Tang
- Department of Acupuncture and Tuina Science, School of Traditional Chinese Medicine, Naval Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China
| | - Cuiling Liu
- Logistics Service Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Weihong Li
- Department of Acupuncture and Tuina Science, School of Traditional Chinese Medicine, Naval Medical University, No. 800 Xiangyin Road, Shanghai, 200433, China.
| |
Collapse
|
13
|
Jia YJ, Li TY, Han P, Chen Y, Pan LJ, Jia CS. Effects of different courses of moxibustion treatment on intestinal flora and inflammation of a rat model of knee osteoarthritis. JOURNAL OF INTEGRATIVE MEDICINE 2022; 20:173-181. [PMID: 35101368 DOI: 10.1016/j.joim.2022.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/15/2021] [Indexed: 01/23/2023]
Abstract
OBJECTIVE This study was done to determine the effects of different courses of moxibustion on a rat knee osteoarthritis (KOA) model, and explore the dose-effect relationship of moxibustion on KOA from the perspectives of intestinal flora and inflammatory factors. METHODS Wistar rats were randomly divided into five groups: normal, model, moxibustion for 2 weeks, moxibustion for 4 weeks and moxibustion for 6 weeks groups (n = 5 each group). A KOA rat model was induced by monosodium iodoacetate, and moxibustion intervention was performed at the acupoints "Dubi" (ST35) and "Zusanli" (ST36), once every other day. Pathologic changes in the cartilage of rat knee joints were assessed after intervention, and fecal samples were subjected to 16S rRNA high-throughput sequencing for microbial diversity analysis. RESULTS Damage to the knee articular cartilage was obvious in the model group, which also had increased levels of pro-inflammatory factors, decreased levels of anti-inflammatory factors, and intestinal flora disorders with decreased diversity. The degree of cartilage damage in the 4 and 6 weeks of moxibustion groups was significantly improved compared with the model group. The 4 and 6 weeks of moxibustion groups also demonstrated reduced levels of interleukin-1β and tumor necrosis factor-α and increased levels of interleukin-10 (P < 0.05). Both the abundance and diversity of the intestinal flora were increased, approaching those of the normal group. Abundances of probiotics Eubacterium coprostanoligenes group and Ruminococcaceae UCG-014 increased, while that of the pathogenic bacteria Lachnospiraceae NK4A136 group decreased (P < 0.05). Although the abundance of Lachnospiraceae NK4A136 group decreased in the 2 weeks of moxibustion group compared with the model group (P < 0.05), there was no statistically significant difference in serum inflammatory factors, flora species diversity or degree of pathological damage compared with the model group. CONCLUSION Moxibustion treatment led to significant improvements in the intestinal flora and inflammatory factors of rats with KOA. Moxibustion treatment of 4 and 6 weeks led to better outcomes than the 2-week course. Moxibustion for 4 and 6 weeks can regulate intestinal flora dysfunction with increased probiotics and reduced pathogenic bacteria, reduce pro-inflammatory factors and increase anti-inflammatory factors. No significant differences were seen between the effects of moxibustion for 4 weeks and 6 weeks.
Collapse
Affiliation(s)
- Ye-Juan Jia
- School of Acupuncture, Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Tian-Yu Li
- School of Acupuncture, Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Peng Han
- School of Acupuncture, Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Yu Chen
- School of Acupuncture, Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Li-Jia Pan
- School of Acupuncture, Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China
| | - Chun-Sheng Jia
- School of Acupuncture, Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang 050200, Hebei Province, China.
| |
Collapse
|
14
|
Yue Y, Chen Y, Liu H, Xu L, Zhou X, Ming H, Chen X, Chen M, Lin Y, Liu L, Zhao Y, Liu S. Shugan Hewei Decoction Alleviates Cecum Mucosal Injury and Improves Depressive- and Anxiety-Like Behaviors in Chronic Stress Model Rats by Regulating Cecal Microbiota and Inhibiting NLRP3 Inflammasome. Front Pharmacol 2022; 12:766474. [PMID: 34987395 PMCID: PMC8721152 DOI: 10.3389/fphar.2021.766474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic stress is a significant cause of depression, anxiety, and intestinal mucosal injury. Gut microbiota disturbances are also associated with these disorders. Shugan Hewei Decoction (SHD), which is a traditional Chinese medicine formula developed by our team, has shown superior therapeutic effects in the treatment of depression, anxiety, and functional gastrointestinal diseases caused by chronic stress. In this study, we investigated the modulatory effect of SHD on the cecal microbiota and cecum mucosal NOD-like receptor protein 3 (NLRP3) inflammasome in a chronic unpredictable stress (CUS)/social isolation rat model. After the SHD intervention, the CUS model rats showed improvements in their depressive- and anxiety-like behaviors, as well as sustained body weight growth and improved fecal characteristics. SHD improved the cecal microbiota diversity and changed the abundance of six microbial genera. A Spearman's correlation analysis showed a strong correlation between the NLRP3 inflammasome and CUS-perturbed cecal biomarker microbiota. SHD regulated the excessive expression of NLRP3, ASC, caspase-1, interleukin-1β (IL-1β), and IL-18 in the serum and cecum mucosa induced by CUS, as well as the activation of the Toll-like receptor 4/nuclear factor-κB signaling cascades. Our results reveal the pharmacological mechanisms of SHD and provide a validated therapeutic method for the treatment of depression, anxiety, and cecum mucosal injury.
Collapse
Affiliation(s)
- Yingying Yue
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China.,Institute of Classical Prescription Applications, Hubei University of Chinese Medicine, Wuhan, China
| | - Yu Chen
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China.,Institute of Classical Prescription Applications, Hubei University of Chinese Medicine, Wuhan, China
| | - Hao Liu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Lesi Xu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China.,Institute of Classical Prescription Applications, Hubei University of Chinese Medicine, Wuhan, China
| | - Xian Zhou
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Hao Ming
- School of Medicine, Jianghan University, Wuhan, China
| | - Xin Chen
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Miaoqi Chen
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yunya Lin
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Lin Liu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Yingqian Zhao
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Songlin Liu
- College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
15
|
Lang W, Cheng M, Zheng X, Zhao Y, Qu Y, Jia Z, Gong H, Ali I, Tang J, Zhang H. Forsythiaside A alleviates methotrexate-induced intestinal mucositis in rats by modulating the NLRP3 signaling pathways. Int Immunopharmacol 2021; 103:108466. [PMID: 34933162 DOI: 10.1016/j.intimp.2021.108466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 11/28/2022]
Abstract
Most chemotherapeutic drugs can kill the tumor cells, but also cause a vast damage to body, such as intestinal mucositis (IM). The present study was design to find out the effect of Forsythiaside A (FTA) on chemotherapeutic-induced IM in rats. Briefly, for 3 consecutive days, male Sprague-Dawley rats were treated with 7 mg / kg methotrexate (MTX) to establish IM and simultaneously administered with 40 or 80 mg / kg FTA for 7 days. Our results showed that the final body weight and daily food intake were increased, and the disease activity index was reduced in the MTX group after FTA treatment. The MTX group showed the pathological alterations like the inflammatory cells infiltration, the mucosal layer destruction, glands expansion, intestinal villi structure disorder and goblet cells reduction, while we found that 80 mg / kg FTA treatment displayed evident reversal effects. ELISA further suggested that TNF-α, IL-1β and IL-18 levels in serum in MTX-induced rats were reduced after 80 mg / kg FTA treatment. Moreover, FTA decreased the number of leukocytes, neutrophils and lymphocytes in peripheral blood. Western blot and immunofluorescence results indicated that the expression levels of NLRP3, cleaved caspase 1, cleaved IL-1β and CD68 positive rate were down-regulated in MTX-induced rats after 80 mg / kg FTA intervention. The findings of the current study suggested that FTA effectively inhibited MTX-induced IM in rats by attenuating the activation of the NLRP3 signaling pathways.
Collapse
Affiliation(s)
- Wuying Lang
- College of Biology Pharmacy and Food Engineering, Shangluo University, Beixin Street 10, Shangluo, China; Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd, Beixin Street 10, Shangluo, China
| | - Min Cheng
- College of Biology Pharmacy and Food Engineering, Shangluo University, Beixin Street 10, Shangluo, China; Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd, Beixin Street 10, Shangluo, China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun, China
| | - Yongping Zhao
- College of Biology Pharmacy and Food Engineering, Shangluo University, Beixin Street 10, Shangluo, China; Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd, Beixin Street 10, Shangluo, China
| | - Yunlong Qu
- College of Biology Pharmacy and Food Engineering, Shangluo University, Beixin Street 10, Shangluo, China; Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd, Beixin Street 10, Shangluo, China
| | - Zhao Jia
- College of Biology Pharmacy and Food Engineering, Shangluo University, Beixin Street 10, Shangluo, China; Shaanxi Qinling Industrial Technology Research Institute of Special Biological Resources Co. Ltd, Beixin Street 10, Shangluo, China
| | - Haizhou Gong
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun, China
| | - Ihsan Ali
- College of veterinary science faculty of animal husbandry and veterinary science, the University of Agriculture Peshawar, Pakistan
| | - Jingwen Tang
- College of Biology Pharmacy and Food Engineering, Shangluo University, Beixin Street 10, Shangluo, China
| | - Haihua Zhang
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (under planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, No. 360 Hebei Street, Haigang District, Qinhuangdao 066004, China.
| |
Collapse
|
16
|
Gupta R, Srivastava D, Sahu M, Tiwari S, Ambasta RK, Kumar P. Artificial intelligence to deep learning: machine intelligence approach for drug discovery. Mol Divers 2021; 25:1315-1360. [PMID: 33844136 PMCID: PMC8040371 DOI: 10.1007/s11030-021-10217-3] [Citation(s) in RCA: 407] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Drug designing and development is an important area of research for pharmaceutical companies and chemical scientists. However, low efficacy, off-target delivery, time consumption, and high cost impose a hurdle and challenges that impact drug design and discovery. Further, complex and big data from genomics, proteomics, microarray data, and clinical trials also impose an obstacle in the drug discovery pipeline. Artificial intelligence and machine learning technology play a crucial role in drug discovery and development. In other words, artificial neural networks and deep learning algorithms have modernized the area. Machine learning and deep learning algorithms have been implemented in several drug discovery processes such as peptide synthesis, structure-based virtual screening, ligand-based virtual screening, toxicity prediction, drug monitoring and release, pharmacophore modeling, quantitative structure-activity relationship, drug repositioning, polypharmacology, and physiochemical activity. Evidence from the past strengthens the implementation of artificial intelligence and deep learning in this field. Moreover, novel data mining, curation, and management techniques provided critical support to recently developed modeling algorithms. In summary, artificial intelligence and deep learning advancements provide an excellent opportunity for rational drug design and discovery process, which will eventually impact mankind. The primary concern associated with drug design and development is time consumption and production cost. Further, inefficiency, inaccurate target delivery, and inappropriate dosage are other hurdles that inhibit the process of drug delivery and development. With advancements in technology, computer-aided drug design integrating artificial intelligence algorithms can eliminate the challenges and hurdles of traditional drug design and development. Artificial intelligence is referred to as superset comprising machine learning, whereas machine learning comprises supervised learning, unsupervised learning, and reinforcement learning. Further, deep learning, a subset of machine learning, has been extensively implemented in drug design and development. The artificial neural network, deep neural network, support vector machines, classification and regression, generative adversarial networks, symbolic learning, and meta-learning are examples of the algorithms applied to the drug design and discovery process. Artificial intelligence has been applied to different areas of drug design and development process, such as from peptide synthesis to molecule design, virtual screening to molecular docking, quantitative structure-activity relationship to drug repositioning, protein misfolding to protein-protein interactions, and molecular pathway identification to polypharmacology. Artificial intelligence principles have been applied to the classification of active and inactive, monitoring drug release, pre-clinical and clinical development, primary and secondary drug screening, biomarker development, pharmaceutical manufacturing, bioactivity identification and physiochemical properties, prediction of toxicity, and identification of mode of action.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Swati Tiwari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|