1
|
Wang Z, Liu Z, Cui L, Sun J, Bu C, Tang M, Li M, Gao S, Chen W, Tao X. Disturbance of bile acids profile aggravates the diarrhea induced by capecitabine through inhibiting the Wnt/β-catenin pathway. J Adv Res 2025; 72:591-604. [PMID: 39048073 DOI: 10.1016/j.jare.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/02/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024] Open
Abstract
INTRODUCTION Diarrhea is the primary dose-limiting side effect of capecitabine(Cap) hindering its clinical application, but the mechanism is unclear. Clarifying this mechanism may enhance the patient compliance and improve the treatment outcome. OBJECTIVES To assess if the endogenous metabolic profile could prodict the diarrhea induced by Cap and explore and validate underlying mechanisms. METHODS Untargeted and targeted bile acids(BAs) metabolomics were performed to analyzed the metabolic profile of baseline samples from colorectal cancer(CRC) patients and the association with the diarrhea induced by Cap was assessed. The toxicity of BAs and Cap and its metabolites alone or their combinations to the human normal intestinal epithelial cell(HIEC) was assessed, and the key genes that mediated the BAs-enhanced toxicity of Cap were discovered by RNA-seq and then validated. A mouse model with high exposure levels of BAs was constructed and then treated with Cap to verify the Cap-induced diarrhea enhanced by BAs. RESULTS The baseline endogenous metabolic profile showed obviously difference between diarrhea and non-diarrhea CRC patients, and the differential metabolites mainly enriched in BAs metabolism; the deoxycholic acid(DCA) and lithocholic acid(LCA) were selected to be the key BAs that enhanced the toxicity of Cap metabolite 5-FU to the HIEC cell; the DCA and LCA could inhibit the Wnt/β-catenin signaling pathway, which then suppressed the P-glycoprotein and increased the exposure level of 5-FU in the HIEC cell. The results of animal experiment verified that the excessive DCA and LCA could aggravate the Cap-induced diarrhea through inhibiting Wnt/β-catenin-P-glycoprotein pathway. CONCLUSIONS The disordered BAs metabolic profile showed close relationship with diarrhea induced by Cap, and excessive DCA and LCA were proved to be the key BAs, which could aggravate the Cap-induced diarrhea through inhibiting Wnt/β-catenin-P-glycoprotein pathway.
Collapse
Affiliation(s)
- Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China
| | - Zhijun Liu
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China; Research and Development Center of Chinese Medicine Resources and Biotechnology, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Lili Cui
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China
| | - Jianguo Sun
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China; College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, 650500, PR China
| | - Chen Bu
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China
| | - Mao Tang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China
| | - Mingming Li
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China; College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, 650500, PR China.
| | - Wansheng Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China; Research and Development Center of Chinese Medicine Resources and Biotechnology, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China.
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, PR China; College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, 650500, PR China.
| |
Collapse
|
2
|
Zhao R, Han M, Lin S, Lin Z, Yu M, Zhang B, Ma L, Li D, Peng L. Adverse drug events associated with fluorouracil use in patients with metastatic colorectal cancer: a real-world pharmacovigilance study based on the FDA adverse event reporting system. Expert Opin Drug Saf 2024; 23:1295-1307. [PMID: 39010662 DOI: 10.1080/14740338.2024.2380513] [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: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Fluorouracil (5-FU) is widely used to treat metastatic colorectal cancer (mCRC), but real-world safety data is limited. Our study aimed to evaluate 5-FU's safety profile in a large mCRC population using the FAERS database. RESEARCH DESIGN AND METHODS We conducted disproportionality analyses to identify adverse drug events associated with 5-FU use in mCRC patients from 2004 to 2023. Subgroup analyses, gender difference analyses, and logistic regression were also performed. RESULTS We identified 1,458 reports with 5-FU as the primary suspected drug, with males accounting for 48.8% of reports. Gastrointestinal disorders were the most common adverse event (864 cases), while pregnancy-related conditions showed the strongest signal intensity (ROR = 2.97). We found 19 preferred terms with positive signals, including ischemic hepatitis (ROR = 59.32), blood iron increased (ROR = 59.32), and stress cardiomyopathy (ROR = 51.94). Males were more susceptible to weight loss and skin toxicity. Most adverse events occurred within the first month of 5-FU administration. CONCLUSION Our study provides a comprehensive analysis of 5-FU's safety profile in mCRC patients, helping healthcare professionals mitigate risks in clinical practice.
Collapse
Affiliation(s)
- Ruiqi Zhao
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengyao Han
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Sen Lin
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Zhimei Lin
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengjiao Yu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Bei Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Lanyue Ma
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Danfei Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Lisheng Peng
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Kuo YR, Lin CH, Lin WS, Pan MH. L-Glutamine Substantially Improves 5-Fluorouracil-Induced Intestinal Mucositis by Modulating Gut Microbiota and Maintaining the Integrity of the Gut Barrier in Mice. Mol Nutr Food Res 2024; 68:e2300704. [PMID: 38656560 DOI: 10.1002/mnfr.202300704] [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: 10/03/2023] [Revised: 03/26/2024] [Indexed: 04/26/2024]
Abstract
SCOPE This study investigates the potential of glutamine to mitigate intestinal mucositis and dysbiosis caused by the chemotherapeutic agent 5-fluorouracil (5-FU). METHODS AND RESULTS Over twelve days, Institute of Cancer Research (ICR) mice are given low (0.5 mg kg-1) or high (2 mg kg-1) doses of L-Glutamine daily, with 5-FU (50 mg kg-1) administered between days six and nine. Mice receiving only 5-FU exhibited weight loss, diarrhea, abnormal cell growth, and colonic inflammation, correlated with decreased mucin proteins, increased endotoxins, reduced fecal short-chain fatty acids, and altered gut microbiota. Glutamine supplementation counteracted these effects by inhibiting the Toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) pathway, modulating nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) oxidative stress proteins, and increasing mammalian target of rapamycin (mTOR) levels, thereby enhancing microbial diversity and protecting intestinal mucosa. CONCLUSIONS These findings underscore glutamine's potential in preventing 5-FU-induced mucositis by modulating gut microbiota and inflammation pathways.
Collapse
Affiliation(s)
- Ya-Ru Kuo
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Cheng-Hung Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
| | - Wei-Sheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
- Department of Food Science, National Quemoy University, Quemoy County, 89250, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung City, 41354, Taiwan
| |
Collapse
|
4
|
Cartwright BM, Corso JN, Lightner J, Whitted C, Torrenegra RD, Krishnan K, Palau VE. Achyrocline B (3,5 dihydroxy-6,7,8-trimethoxyflavone) synergizes with 5-fluorouracil allowing for dose reduction and reduced off-target toxicity in the treatment of colonic and pancreatic cancers. Biomed Pharmacother 2023; 167:115546. [PMID: 37741250 DOI: 10.1016/j.biopha.2023.115546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Surgically unresectable colorectal and pancreatic carcinomas have a high rate of mortality as current therapeutic options are limited. One common chemotherapeutic used to broadly treat both cancers is 5-flurouracil (5-Fu); however, treatment serves only to slow progression of the disease and comes with many side effects due to 5-Fu's intrinsic toxicity. Thus, strategies to decrease the dose of 5-Fu utilized therapeutically as well as reduce 5-Fu's off-target toxicity are paramount. Using cell models of colorectal and pancreatic cancers, we show that cotreatment with Achyrocline B (3,5 dihydroxy-6,7,8-trimethoxyflavone, AcB), a natural flavone from Achyrocline bogotensis, allows for four-fold reduction in 5-Fu dosage without loss of efficacy. We further show that the action of AcB is due to continued cell cycle progression despite 5-Fu pressure to synchronize at the G1/S threshold. In addition to AcB's effect on cancer cells, we found that AcB can directly reduce toxicity of 5-Fu in cells mimicking non-cancerous tissues. These in vitro results are then supported by xenograft modeling. AcB was shown to increase apoptosis in tumors leading to degeneration of the outer tumoral boundary. Furthermore, in 5-Fu treated animals it was found that AcB provided protection to the intestinal tract as indicated by preserved histological and immunohistochemical features. These results show promise for a new adjuvant therapy for colorectal and pancreatic carcinomas that not only reduces tumor progression, but more importantly has the potential to improve patient quality of life.
Collapse
Affiliation(s)
- Brian M Cartwright
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, 37614, United States; Department of Pathology, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Jaclyn N Corso
- Department of Internal Medicine, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Janet Lightner
- Department of Internal Medicine, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Crystal Whitted
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, 37614, United States
| | - Ruben D Torrenegra
- Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales, Bogota, Colombia
| | - Koyamangalath Krishnan
- Department of Internal Medicine, ETSU Quillen College of Medicine, Johnson City, TN, 37614, United States
| | - Victoria E Palau
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, 37614, United States; Productos Naturales, Universidad de Ciencias Aplicadas y Ambientales, Bogota, Colombia.
| |
Collapse
|
5
|
Kawasaki Y, Kakimoto K, Tanaka Y, Shimizu H, Nishida K, Numa K, Kinoshita N, Tatsumi Y, Nakazawa K, Koshiba R, Hirata Y, Ota K, Sakiyama N, Terazawa T, Takeuchi T, Miyazaki T, Goto M, Yokota H, Makizaki Y, Tanaka Y, Nakajima S, Ohno H, Higuchi K, Nakamura S, Nishikawa H. Relationship between Chemotherapy-Induced Diarrhea and Intestinal Microbiome Composition. Digestion 2023; 104:357-369. [PMID: 37231829 PMCID: PMC10614279 DOI: 10.1159/000528282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/31/2022] [Indexed: 05/27/2023]
Abstract
BACKGROUND AND AIM Fluoropyrimidines (FPs) are key drugs in many chemotherapy regimens; however, recipients are often prone to diarrhea due to gastrointestinal toxicity. Disruption of the intestinal epithelial barrier function by FPs leads to dysbiosis, which may exacerbate intestinal epithelial cell damage as a secondary effect and trigger diarrhea. However, despite studies on chemotherapy-induced changes in the intestinal microbiome of humans, the relationship between dysbiosis and diarrhea is unclear. In this study, we aimed to investigate the relationship between chemotherapy-induced diarrhea and the intestinal microbiome. METHODS We conducted a single-center prospective observational study. Twenty-three patients who received chemotherapy, including FPs as first-line chemotherapy for colorectal cancer, were included. Stool samples were collected before the start of chemotherapy and after one cycle of treatment to analyze intestinal microbiome composition and perform PICRUSt predictive metagenomic analysis. RESULTS Gastrointestinal toxicity was observed in 7 of 23 patients (30.4%), diarrhea was observed in 4 (17.4%), and nausea and anorexia were observed in 3 (13.0%). In 19 patients treated with oral FPs, the α diversity of the microbial community decreased significantly following chemotherapy only in the diarrheal group. At the phylum level, the diarrheal group showed a significant decrease in the abundance of Firmicutes and a significant increase in the abundance of Bacteroidetes with chemotherapy (p = 0.013 and 0.011, respectively). In the same groups, at the genus level, Bifidobacterium abundance was significantly decreased (p = 0.019). In contrast, in the non-diarrheal group, Actinobacteria abundance increased significantly with chemotherapy at the phylum level (p = 0.011). Further, Bifidobacterium, Fusicatenibacter, and Dorea abundance significantly increased at the genus level (p = 0.006, 0.019, and 0.011, respectively). The PICRUSt predictive metagenomic analysis revealed that chemotherapy caused significant differences in membrane transport in KEGG pathway level 2 and in 8 KEGG pathway level 3, including transporters and oxidative phosphorylation in the diarrhea group. CONCLUSION Organic-acid-producing bacteria seem to be involved in diarrhea associated with chemotherapy, including FPs.
Collapse
Affiliation(s)
- Yuka Kawasaki
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kazuki Kakimoto
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yasuyoshi Tanaka
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Hikaru Shimizu
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Koji Nishida
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Keijiro Numa
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Naohiko Kinoshita
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yoshihiro Tatsumi
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kei Nakazawa
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Ryoji Koshiba
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Yuki Hirata
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kazuhiro Ota
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Naokuni Sakiyama
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Tetsuji Terazawa
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Toshihisa Takeuchi
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Takako Miyazaki
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Masahiro Goto
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Haruka Yokota
- R&D Center, Biofermin Pharmaceutical Co., Ltd., Kobe, Japan
| | | | - Yoshiki Tanaka
- R&D Center, Biofermin Pharmaceutical Co., Ltd., Kobe, Japan
| | | | - Hiroshi Ohno
- R&D Center, Biofermin Pharmaceutical Co., Ltd., Kobe, Japan
| | - Kazuhide Higuchi
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shiro Nakamura
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Hiroki Nishikawa
- 2nd Department of Internal Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
6
|
Mohammed AI, Celentano A, Paolini R, Low JT, McCullough MJ, O' Reilly LA, Cirillo N. Characterization of a novel dual murine model of chemotherapy-induced oral and intestinal mucositis. Sci Rep 2023; 13:1396. [PMID: 36697446 PMCID: PMC9876945 DOI: 10.1038/s41598-023-28486-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Oral and intestinal mucositis are debilitating inflammatory diseases observed in cancer patients undergoing chemo-radiotherapy. These are devastating clinical conditions which often lead to treatment disruption affecting underlying malignancy management. Although alimentary tract mucositis involves the entire gastrointestinal tract, oral and intestinal mucositis are often studied independently utilizing distinct organ-specific pre-clinical models. This approach has however hindered the development of potentially effective whole-patient treatment strategies. We now characterize a murine model of alimentary tract mucositis using 5-Fluorouracil (5-FU). Mice were given 5-FU intravenously (50 mg/kg) or saline every 48 h for 2 weeks. Post initial injection, mice were monitored clinically for weight loss and diarrhea. The incidence and extent of oral mucositis was assessed macroscopically. Microscopical and histomorphometric analyses of the tongue and intestinal tissues were conducted at 3 interim time points during the experimental period. Repeated 5-FU treatment caused severe oral and intestinal atrophy, including morphological damage, accompanied by body weight loss and mild to moderate diarrhea in up to 77.8% of mice. Oral mucositis was clinically evident throughout the observation period in 88.98% of mice. Toluidine blue staining of the tongue revealed that the ulcer size peaked at day-14. In summary, we have developed a model reproducing the clinical and histologic features of both oral and intestinal mucositis, which may represent a useful in vivo pre-clinical model for the study of chemotherapy-induced alimentary tract mucositis and the development of preventative therapies.
Collapse
Affiliation(s)
- Ali I Mohammed
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia. .,College of Dentistry, The University of Tikrit, Tikrit, Iraq.
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Jun T Low
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Michael J McCullough
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Lorraine A O' Reilly
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3000, Australia
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia.
| |
Collapse
|
7
|
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
|
8
|
Xu C, Shang Z, Najafi M. Lung Pneumonitis and Fibrosis in Cancer Therapy: A Review on Cellular and Molecular Mechanisms. Curr Drug Targets 2022; 23:1505-1525. [PMID: 36082868 DOI: 10.2174/1389450123666220907144131] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/05/2022] [Accepted: 08/02/2022] [Indexed: 01/25/2023]
Abstract
Fibrosis and pneumonitis are the most important side effects of lung tissue following cancer therapy. Radiotherapy and chemotherapy by some drugs, such as bleomycin, can induce pneumonitis and fibrosis. Targeted therapy and immunotherapy also may induce pneumonitis and fibrosis to a lesser extent compared to chemotherapy and radiotherapy. Activation of lymphocytes by immunotherapy or infiltration of inflammatory cells such as macrophages, lymphocytes, neutrophils, and mast cells following chemo/radiation therapy can induce pneumonitis. Furthermore, the polarization of macrophages toward M2 cells and the release of anti-inflammatory cytokines stimulate fibrosis. Lung fibrosis and pneumonitis may also be potentiated by some other changes such as epithelial-mesenchymal transition (EMT), oxidative stress, reduction/oxidation (redox) responses, renin-angiotensin system, and the upregulation of some inflammatory mediators such as a nuclear factor of kappa B (NF-κB), inflammasome, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS). Damages to the lung vascular system and the induction of hypoxia also can induce pulmonary injury following chemo/radiation therapy. This review explains various mechanisms of the induction of pneumonitis and lung fibrosis following cancer therapy. Furthermore, the targets and promising agents to mitigate lung fibrosis and pneumonitis will be discussed.
Collapse
Affiliation(s)
- Chaofeng Xu
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China
| | - Zhongtu Shang
- Zhuji People's Hospital of Zhejiang Province, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang, 311800, China
| | - Masoud Najafi
- Medical Technology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
9
|
Simonyan RM, Simonyan KV, Simonyan GM, Khachatryan HS, Babayan MA, Danielyan MH, Darbinyan LV, Simonyan MA. Superoxide-producing thermostable associate from the small intestines of control and alloxan-induced diabetic rats: quantitative and qualitative changes. BMC Endocr Disord 2022; 22:250. [PMID: 36258207 PMCID: PMC9580182 DOI: 10.1186/s12902-022-01160-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/28/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND NADPH oxidase 1 (Nox1), which is highly expressed in the colon, is thought to play a potential role in host defense as a physical and innate immune barrier against commensal or pathogenic microbes in the gastrointestinal epithelium. Diabetes can be caused by several biological factors, including insulin resistance is one of them. Alloxan is widely used to induce insulin-dependent diabetes in experimental animals. Alloxan increases the generation of reactive oxygen species as a result of metabolic reactions in the body, along with a massive increase in cytosolic calcium concentration. METHODS Using a universal method, a superoxide radical (О2-)-thermostable associate between NADPH-containing lipoprotein (NLP) and NADPH oxidase (Nox)- NLP-Nox was isolated and purified from the small intestine (SI) of control (C) and alloxan-induced diabetic (AD) albino rats. RESULTS In comparison to the C indices, in AD in the SI, an increase in the specific content of NLP-Nox associate and a decrease in the stationary concentration of produced О2- in liquid phase (in solution) and gas phase (during blowing by oxygen of the NLP-Nox solution) were observed. The NLP-Nox of SI associate in C and AD rats produced О2- by an immediate mechanism, using NLP as a substrate. The phenomenon of the hiding of the optical absorption maxima of the Nox in oxidized states at pH10,5 was observed in the composition of these SI associates of the C and AD rat groups. The characteristic absorption maxima of the «hidden» Nox were observed under these conditions after reduction by potassium dithionite. CONCLUSION Thus, at AD, the decrease in the stationary concentration of produced О2- in the solution and gas phase was compensated for by an increase in the specific amount of associate. In addition, the decrease in the stationary concentration of produced О2- by NLP-Nox associates at AD can be linked to a decrease in the level of NADPH in NLP-Nox composition. This could be used as a new mechanism of AD pathogenesis.
Collapse
Affiliation(s)
- R M Simonyan
- H. Buniatyan Institute of Biochemistry NAS RA, 0014, Yerevan, Armenia
| | - K V Simonyan
- Neuroendocrine Relationships Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia.
| | - G M Simonyan
- H. Buniatyan Institute of Biochemistry NAS RA, 0014, Yerevan, Armenia
| | - H S Khachatryan
- H. Buniatyan Institute of Biochemistry NAS RA, 0014, Yerevan, Armenia
| | - M A Babayan
- H. Buniatyan Institute of Biochemistry NAS RA, 0014, Yerevan, Armenia
| | - M H Danielyan
- Histochemistry and Electron microscopy Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - L V Darbinyan
- Sensorimotor Integration Lab, Orbeli Institute of Physiology NAS RA, 0028, Yerevan, Armenia
| | - M A Simonyan
- H. Buniatyan Institute of Biochemistry NAS RA, 0014, Yerevan, Armenia
| |
Collapse
|
10
|
Xia F, Li Y, Deng L, Ren R, Ge B, Liao Z, Xiang S, Zhou B. Alisol B 23-Acetate Ameliorates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction by Inhibiting TLR4-NOX1/ROS Signaling Pathway in Caco-2 Cells. Front Pharmacol 2022; 13:911196. [PMID: 35774596 PMCID: PMC9237229 DOI: 10.3389/fphar.2022.911196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/24/2022] [Indexed: 01/13/2023] Open
Abstract
Alisol B 23-Acetate (AB23A) is a naturally occurring triterpenoid, which can be indicated in the rhizome of medicinal and dietary plants from Alisma species. Previous studies have demonstrated that AB23A could inhibit intestinal permeability by regulating tight junction (TJ)-related proteins. Even so, the AB23A protective mechanism against intestinal barrier dysfunction remains poorly understood. This investigation seeks to evaluate the AB23A protective effects on intestinal barrier dysfunction and determine the mechanisms for restoring intestinal barrier dysfunction in LPS-stimulated Caco-2 monolayers. According to our findings, AB23A attenuated the inflammation by reducing pro-inflammatory cytokines production like IL-6, TNF-α, IL-1β, and prevented the paracellular permeability by inhibiting the disruption of TJ in LPS-induced Caco-2 monolayers after treated with LPS. AB23A also inhibited LPS-induced TLR4, NOX1 overexpression and subsequent ROS generation in Caco-2 monolayers. Transfected with NOX1-specific shRNA diminished the up-regulating AB23A effect on ZO-1 and occludin expression. Moreover, transfected with shRNA of TLR4 not only enhanced ZO-1 and occludin expression but attenuated NOX1 expression and ROS generation. Therefore, AB23A ameliorates LPS-induced intestinal barrier dysfunction by inhibiting TLR4-NOX1/ROS signaling pathway in Caco-2 monolayers, suggesting that AB23A may have positive impact on maintaining the intestinal barrier’s integrity.
Collapse
Affiliation(s)
- Fan Xia
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Fan Xia, ; Benjie Zhou,
| | - Yuxin Li
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, China
| | - Lijun Deng
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Ruxia Ren
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bingchen Ge
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Ziqiong Liao
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Benjie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Fan Xia, ; Benjie Zhou,
| |
Collapse
|
11
|
Ji Y, Zhou W, Tan W, Chen Z, Lu H, You Y, Tian C, Zhou X, Zhou L, Luo R, Zhao X. Protective effect of polysaccharides isolated from the seeds of Cuscuta chinensis Lam. on 5-fluorouracil-induced intestinal mucositis in mice. Acta Cir Bras 2022; 37:e370204. [PMID: 35507968 PMCID: PMC9064182 DOI: 10.1590/acb370204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 01/10/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose: To evaluate the protective effect of Cuscuta chinensis Lam. polysaccharides (PCCL) on 5-fluorouracil-(5-FU)-induced intestinal mucositis (IM) in mice. Methods: PCCL was orally administered at a dose of 20 mg·kg–1 for 7 days and its protective effect on 5-FU-induced IM (5-FU, 50 mg·kg–1 for 5 days) was evaluated by monitoring changes in body weight, degree of diarrhea, levels of tissue inflammatory factors (tumor necrosis factor α, interleukin 6, and interleukin 1β levels), apoptosis rates, and the expression levels of caspase-3, Bax and Bcl-2. Results: The severity of mucosal injury (as reflected by body weight changes, degree of diarrhea, height of villi, and damage to crypts) was significantly attenuated by PCCL administration. PCCL also reduced the levels of tissue inflammatory factors, the apoptosis rate, and the expression of caspase-3 and Bax, and increased Bcl-2 expression. Conclusions: PCCL administration may be significantly protective against 5-FU-induced IM by inhibiting apoptosis and regulating the abnormal inflammation associated with it.
Collapse
Affiliation(s)
- Yanzhao Ji
- Shanxi Academy of Medical Sciences, China
| | | | - Wei Tan
- Guangdong Academy of Medical Sciences, China
| | | | - Hanqi Lu
- Southern Medical University, China
| | | | | | | | - Lin Zhou
- Southern Medical University, China
| | - Ren Luo
- Southern Medical University, China
| | | |
Collapse
|
12
|
Chukwunyere U, Mercan M, Sehirli AO, Abacioglu N. Possible cytoprotective mechanisms of oxytocin against 5-fluorouracil-induced gastrointestinal mucositis. Mol Biol Rep 2022; 49:4055-4059. [PMID: 35474056 DOI: 10.1007/s11033-022-07384-3] [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: 02/03/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/27/2022]
Abstract
Gastrointestinal mucositis is a common and dose-limiting side effect characterized by ulcerative lesions in the mucosa of the digestive tract in patients receiving anticancer drugs such as 5-fluorouracil (5-FU), a potent antineoplastic drug. Several protocols have reported the efficacy of therapeutic interventions to prevent this side effect, although complete success has not yet been achieved and mucositis remains one of the most serious complications associated with 5-FU therapy. Oxytocin, a well-known antistress agent, has been reported to have comparable effects to ranitidine. Previous studies have shown that oxytocin inhibits gastric acid secretion and the expression of proinflammatory cytokines in rats. If oxytocin can reduce stress-induced ulcers via antioxidant, antiapoptotic, and anti-inflammatory pathways, then it may have a dose-dependent effect on gastrointestinal mucositis caused by 5-FU.
Collapse
Affiliation(s)
- Ugochukwu Chukwunyere
- Department of Pharmacology, Faculty of Pharmacy, Near East University, 99138, Lefkosa, TRNC, Turkey.
| | - Merve Mercan
- Department of Pharmacology, Faculty of Pharmacy, Near East University, 99138, Lefkosa, TRNC, Turkey
| | - Ahmet Ozer Sehirli
- Department of Pharmacology, Faculty of Dentistry, Near East University, Lefkosa, TRNC, Turkey
| | - Nurettin Abacioglu
- Department of Pharmacology, Faculty of Pharmacy, Near East University, 99138, Lefkosa, TRNC, Turkey
| |
Collapse
|
13
|
Yoneda J, Nishikawa S, Kurihara S. Oral administration of cystine and theanine attenuates 5-fluorouracil-induced intestinal mucositis and diarrhea by suppressing both glutathione level decrease and ROS production in the small intestine of mucositis mouse model. BMC Cancer 2021; 21:1343. [PMID: 34922485 PMCID: PMC8684148 DOI: 10.1186/s12885-021-09057-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chemotherapy is frequently used in cancer treatment; however, it may cause adverse events, which must be managed. Reactive oxygen species (ROS) have been reported to be involved in the induction of intestinal mucositis and diarrhea, which are common side effects of treatment with fluoropyrimidine 5-fluorouracil (5-FU). Our previous studies have shown that oral administration of cystine and theanine (CT) increases glutathione (GSH) production in vivo. In the present study, we hypothesized that CT might inhibit oxidative stress, including the overproduction of ROS, and attenuate 5-FU-induced mucositis and diarrhea. METHODS We investigated the inhibitory effect of CT administration on mucositis and diarrhea, as well as its mechanism, using a mouse model of 5-FU-induced intestinal mucositis. RESULTS CT administration suppressed 5-FU-induced diarrhea and weight loss in the studied mice. After 5-FU administration, the GSH level and the GSH/GSSG ratio in the small intestine mucosal tissue decreased compared to normal control group; but CT administration improved the GSH/GSSG ratio to normal control levels. 5-FU induced ROS production in the basal region of the crypt of the small intestine mucosal tissue, which was inhibited by CT. CT did not affect the antitumor effect of 5-FU. CONCLUSIONS CT administration suppressed intestinal mucositis and diarrhea in a mouse model. This finding might be associated with the antioxidant characteristics of CT, including the improved rate of GSH redox and the reduced rate of ROS production in the small intestine mucosal tissue. CT might be a suitable candidate for the treatment of gastrointestinal mucositis associated with chemotherapy.
Collapse
Affiliation(s)
- Junya Yoneda
- Research Institute For Bioscience Products & Fine Chemicals Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, 210-8681, Japan.
| | - Sachiko Nishikawa
- Research Institute For Bioscience Products & Fine Chemicals Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, 210-8681, Japan
| | - Shigekazu Kurihara
- Research Institute For Bioscience Products & Fine Chemicals Ajinomoto Co., Inc., 1-1, Suzuki-Cho, Kawasaki-ku, Kawasaki-shi, 210-8681, Japan
| |
Collapse
|
14
|
Mohamed MZ, Mohammed HH. Rupatadine protects the intestinal mucosa from injury by 5-flurouracil via modulation of inflammation, apoptosis and intestinal permeability. Drug Chem Toxicol 2021; 45:2843-2851. [PMID: 34747284 DOI: 10.1080/01480545.2021.1997541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Fluorouracil (5-FU) is a widely used chemotherapeutic agent in various malignant tumors. However, intestinal toxicity is considered the irritant unavoidable adverse effect during the course therapy. The aim of the current study was to screen the effect of a new selective histamine receptor 1 blocker and platelet-activating factor (PAF) blocker on 5-FU induced intestinal toxicity. Five groups (6 rats each) of adult male rats (Wistar) were arranged as follows: (1) control group that was treated with carboxymethylcellulose, (2) a group that received rupatadine (higher dose) only, (3) a group that received 5-FU and (4) and (5) groups that received 5-FU plus lower or higher dose rupatadine, respectively. At end of the experiment, we determined intestinal malondialdehyde (MDA), glutathione reduced (GSH), nitric oxide (NO), tumor necrosis factor (TNF-α), interleukin 1β, 6, 10 (IL-1β, IL-6, IL-10), PAF, histamine, myeloperoxidase, cysteine-aspartic acid protease-3 (caspase-3), and nuclear factor kappa B (NF-κB) as well as the histological analysis. 5-FU injection caused marked elevation of MDA, NO, TNF-α, IL-1β, IL-6, PAF, histamine, myeloperoxidase, caspase-3, and NF-κB expressions. The intoxicated animals showed deficient GSH and IL-10 along with significant loss of villi, disorganized crypts, and inflammatory cell infiltration. Rupatadine pretreatment reduced the previously mentioned parameters, preserved a nearly normal intestinal mucosa picture with replenished GSH and elevated IL-10. In conclusion, rupatadine is a dual histamine receptor 1, and a PAF blocker could reduce 5-FU-induced oxidative damage, inflammation, apoptosis, and ulceration of the intestinal epithelium. Rupatadine may be a valuable modality to decrease 5-FU induced intestinal mucositis.
Collapse
Affiliation(s)
- Mervat Z Mohamed
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Hanaa H Mohammed
- Department of Histology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
15
|
Pin C, Collins T, Gibbs M, Kimko H. Systems Modeling to Quantify Safety Risks in Early Drug Development: Using Bifurcation Analysis and Agent-Based Modeling as Examples. AAPS JOURNAL 2021; 23:77. [PMID: 34018069 PMCID: PMC8137611 DOI: 10.1208/s12248-021-00580-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 03/09/2021] [Indexed: 11/30/2022]
Abstract
Quantitative Systems Toxicology (QST) models, recapitulating pharmacokinetics and mechanism of action together with the organic response at multiple levels of biological organization, can provide predictions on the magnitude of injury and recovery dynamics to support study design and decision-making during drug development. Here, we highlight the application of QST models to predict toxicities of cancer treatments, such as cytopenia(s) and gastrointestinal adverse effects, where narrow therapeutic indexes need to be actively managed. The importance of bifurcation analysis is demonstrated in QST models of hematologic toxicity to understand how different regions of the parameter space generate different behaviors following cancer treatment, which results in asymptotically stable predictions, yet highly irregular for specific schedules, or oscillating predictions of blood cell levels. In addition, an agent-based model of the intestinal crypt was used to simulate how the spatial location of the injury within the crypt affects the villus disruption severity. We discuss the value of QST modeling approaches to support drug development and how they align with technological advances impacting trial design including patient selection, dose/regimen selection, and ultimately patient safety.
Collapse
Affiliation(s)
- Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge Science Park, Milton Road, Cambridge, UK
| | - Teresa Collins
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge Science Park, Milton Road, Cambridge, UK
| | - Megan Gibbs
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Holly Kimko
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gaithersburg, Maryland, USA.
| |
Collapse
|
16
|
Sougiannis AT, VanderVeen BN, Davis JM, Fan D, Murphy EA. Understanding chemotherapy-induced intestinal mucositis and strategies to improve gut resilience. Am J Physiol Gastrointest Liver Physiol 2021; 320:G712-G719. [PMID: 33471628 PMCID: PMC8202195 DOI: 10.1152/ajpgi.00380.2020] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal mucositis remains one of the most debilitating side effects related to chemotherapy. The onset and persistence of mucositis is an intricate physiological process involving cross-communication between the specific chemotherapeutic drug, the immune system, and gut microbes that results in a loss of mucosal integrity leading to gut-barrier dysfunction. Intestinal mucositis has a severe impact on a patient's quality of life and negatively influences the outcome of treatment. Most importantly, intestinal mucositis is a major contributor to the decreased survival rates and early onset of death associated with certain chemotherapy treatments. Understanding the pathophysiology and symptomology of intestinal mucositis is important in reducing the negative consequences of this condition. Prophylaxis, early diagnosis, and proper symptom management are essential to improved survival outcomes in patients with cancer. This review focuses on the pathobiology of intestinal mucositis that accompanies chemotherapy treatments. In addition, we will discuss the therapeutic potential of select strategies that have shown promise in mitigating chemotherapies' off-target effects without hampering their anticancer efficacy.NEW & NOTEWORTHY Intestinal mucositis, or damage to the intestinal mucosa, is a common side effect of chemotherapy. In this review, we describe the pathobiology of intestinal mucositis that is associated with chemotherapy treatments. In addition, we discuss the efficacy of several potential therapeutic strategies that have shown some potential in alleviating chemotherapies' off-target effects.
Collapse
Affiliation(s)
- Alexander T. Sougiannis
- 1Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, South Carolina,2College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Brandon N. VanderVeen
- 1Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, South Carolina,3AcePre, LLC, Columbia, South Carolina
| | - J. Mark Davis
- 3AcePre, LLC, Columbia, South Carolina,4Department of Exercise Science, Arnold School of Public Health, University of South Carolina, South Carolina
| | - Daping Fan
- 3AcePre, LLC, Columbia, South Carolina,5Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, South Carolina
| | - E. Angela Murphy
- 1Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, South Carolina,3AcePre, LLC, Columbia, South Carolina
| |
Collapse
|
17
|
Abdellateif MS, Salem SE, Badr DM, Shaarawy S, Hussein MM, Zekri ARN, Fouad MA. The Prognostic Significance of 5-Fluorouracil Induced Inflammation and Immuno-Modulation in Colorectal Cancer Patients. J Inflamm Res 2020; 13:1245-1259. [PMID: 33408498 PMCID: PMC7781028 DOI: 10.2147/jir.s283069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Aim The change in the levels of peripheral inflammatory markers together with EGFR in relation to 5- fluorouracil (5-FU) therapy was evaluated for their prognostic significance in colorectal cancer (CRC) patients. Patients and Methods Expression levels of COX2, IL6, IL1β, EGFR, IL10, and TNFα were determined with quantitative real-time PCR (qPCR) in the peripheral blood of 90 CRC patients. The inflammatory response was correlated with patients’ clinical features, disease-free survival (DFS), and overall survival (OS). Results After 6 months of 5-FU therapy, increased inflammatory response was found to be associated with smoking, T3 or T4 tumors, performance status (PS) III, positive lymph nodes, distant metastasis, and gastrointestinal (GIT) toxicity. The combination of COX2 with interleukins in a predictive equation for DFS was significant in patients with over-expression of EGFR. DFS and OS rates were reduced in patients with increased COX2, IL6, IL10, and TNFα expression with 5-FU therapy. Significant hazard of disease progression was associated with smoking (HR=1.27, P=0.004), 5-FU induction of COX2, and IL6 expression (HR=1.35, P=0.001 and HR=1.27, P=0.004, respectively). Moreover, smoking, 5-FU induction of IL6, TNFα, and IL10 expression are found to be independent prognostic factors for OS (P=0.003, 0.003, 0.002, and 0.002, respectively). Conclusion The peripheral effects of 5-FU therapy have shown a significant impact on the treatment outcome of CRC patients.
Collapse
Affiliation(s)
- Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Salem E Salem
- Medical Oncology Department, National Cancer Institute, Cairo, Egypt
| | - Doaa M Badr
- Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sabry Shaarawy
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Marwa M Hussein
- Medical Oncology Department, National Cancer Institute, Cairo, Egypt
| | - Abdel-Rahman N Zekri
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo, Egypt
| | - Mariam A Fouad
- Pharmacology and Experimental Oncology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
18
|
Kamar SS, Baky MH, Omar AI. The beneficial influence of rhubarb on 5-fluorouracil-induced ileal mucositis and the combined role of aquaporin-4, tumour necrosis factor-α, nuclear factor-kappa B & matrix metalloproteinase-9 in rat model: histological study. Anat Cell Biol 2020; 53:228-239. [PMID: 32647090 PMCID: PMC7343559 DOI: 10.5115/acb.20.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 11/27/2022] Open
Abstract
A 5-fluorouracil (5-FU) is used for cancer treatment despite its cytotoxic sequelae on healthy cells, especially the rapid proliferating ones. Intestinal mucositis is one of the most frequent chemotherapeutic debilitating sequelae. Rhubarb (Rh), an ancient herb, is known for its curing effect on gastrointestinal complications. This study aims to detect the role of aquaporin-4 (AQP-4), tumour necrosis factor-α (TNF-α), nuclear factor-kappa B (NF-κB), and matrix metalloproteinase-9 (MMP-9) in 5-FU-induced ileal histological and biochemical changes and the potential therapeutic effect of Rh water extract on these changes in rats. A 45 rats were divided into 3 groups; control, 5-FU (single intraperitoneal injection of 150 mg/kg/rat) and Rh-treated (oral 20 mg/kg/day/rat for 8 days). The change in animals’ weight, incidence of diarrhoea and AQP-4 and TNF-α values in ileal homogenates were measured. Ileal sections were subjected to hematoxylin and eosin stain, periodic acid Schiff (PAS)-reaction and MMP-9, NF-κB and AQP-4 immunohistochemical staining. A 5-FU group revealed marked ileal mucosal damage associated with a significant decrease in the mean body weight, AQP-4 level and area percent of PAS and AQP-4 positive reaction. Significant increase in the mean incidence of diarrhoea, TNF-α value and area percent of MMP-9 and NF-κB was detected. These changes were significantly corrected with Rh administration. A 5-FU resulted in severe ileal mucositis through TNF-α, NF-κB, MMP-9, and AQP-4 disturbances. Rh treatment was highly effective in preventing such mucositis.
Collapse
Affiliation(s)
- Samaa Samir Kamar
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mostafa Hasan Baky
- Department of Pharmacology, Faculty of Pharmacology, Egyptian Russian University, Cairo, Egypt
| | - Abeer Ibraheem Omar
- Department of Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
19
|
Qu L, Tan W, Yang J, Lai L, Liu S, Wu J, Zou W. Combination Compositions Composed of l-Glutamine and Si-Jun-Zi-Tang Might Be a Preferable Choice for 5-Fluorouracil-Induced Intestinal Mucositis: An Exploration in a Mouse Model. Front Pharmacol 2020; 11:918. [PMID: 32625099 PMCID: PMC7313676 DOI: 10.3389/fphar.2020.00918] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 06/05/2020] [Indexed: 12/30/2022] Open
Abstract
Intestinal mucositis is a common toxicity of many anti-neoplastic therapies that negatively influences health, the quality of life, economic outcomes, and even the success of cancer treatment. Unfortunately, there is presently no optimal medicine that is able to effectively manage this condition. l-glutamine is one of the most frequently used agent in practice among the limited treatment choices due to its safety and inexpensiveness despite there being a lack of evidence. Previous studies indicated that l-glutamine may alleviate mucositis and mucosal atrophy but failed to improve patients' macroscopic conditions, such as the occurrence of diarrhea. A compound glutamine capsule (G-SJZ), composed of l-glutamine and the traditional Chinese herbal formula Si-Jun-Zi-Tang, has been used in China for 23 years to treat many types of gastrointestinal diseases, including gastrointestinal reactions induced by radiotherapy and chemotherapy. However, the exact effect of G-SJZ on intestinal mucositis is unclear, and moreover, whether l-glutamine combined with Si-Jun-Zi-Tang is more effective than l-glutamine alone have not been studied. In the current study, we explored the effects of G-SJZ and l-glutamine in a mouse model of intestinal mucositis induced by 5-fluorouracil (5-Fu). The results revealed that pretreatment with G-SJZ ameliorated the physical manifestations of weight loss and the severity of diarrhea following continuous 5-Fu injections in mice. Likewise, the histopathological damage and the destruction of villus and crypt structures in the intestinal mucosa as well as the increase in circulating intestinal injury markers caused by 5-Fu were reversed with G-SJZ pretreatment. Furthermore, the protective effect of G-SJZ was accompanied by modulations in the immunohistochemical expression of tight junction proteins. Interestingly, although treatment with a dose of l-glutamine alone that was equivalent to the dose in G-SJZ also showed a protective effect, it did not appear to be as strong as treatment with G-SJZ. Si-Jun-Zi-Tang in G-SJZ may compensate for the deficiencies of l-glutamine in this model which seems not to be related to the regulation of tight junction proteins. Our study is the first to suggest that the combined use of l-glutamine and Si-Jun-Zi-Tang might be more effective than l-glutamine alone despite exact mechanism still needs further study. Because of the limited number of therapeutic agents, G-SJZ is likely to be a preferable choice for intestinal mucositis.
Collapse
Affiliation(s)
- Liping Qu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wanxian Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Limin Lai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sili Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Wenjun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
20
|
Liu J, Iwata K, Zhu K, Matsumoto M, Matsumoto K, Asaoka N, Zhang X, Ibi M, Katsuyama M, Tsutsui M, Kato S, Yabe-Nishimura C. NOX1/NADPH oxidase in bone marrow-derived cells modulates intestinal barrier function. Free Radic Biol Med 2020; 147:90-101. [PMID: 31838229 DOI: 10.1016/j.freeradbiomed.2019.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 01/02/2023]
Abstract
The involvement of reactive oxygen species (ROS) has been suggested in the development of inflammatory bowel disease (IBD). An impaired intestinal barrier function is common in IBD patients. Here, we report the central role of NOX1/NADPH oxidase, a major source of ROS in nonphagocytic cells, in intestinal barrier dysfunction. By in vivo imaging using L-012 as a probe, a time-dependent increase in ROS was demonstrated in the abdomen of wild-type mice (WT) administered lipopolysaccharide (LPS: 6 mg/kg i.p.), but it was almost completely abolished in mice deficient in Nox1 (Nox1-KO) or the inducible nitric oxide synthase gene (iNOS-KO). By ex vivo imaging, increased ROS production was mainly shown in the ileum, where enhanced immunostaining of NOX1 was observed on the apical side of the epithelium. On the other hand, a punctate staining pattern of 3-nitrotyrosine, a marker of peroxynitrite production, was demonstrated in the lamina propria. When LPS-induced intestinal hyperpermeability was assessed by the oral administration of fluorescein isothiocyanate-conjugated dextran (FD-4), it was significantly suppressed in Nox1-KO as well as iNOS-KO. When Nox1-KO adoptively transferred with WT bone marrow were treated with LPS, the serum level of FD-4 was significantly elevated, whereas it remained unchanged in WT receiving bone marrow derived from Nox1-KO. Concomitantly, the activation of matrix metalloproteinase-9 induced by LPS was alleviated not only in intestinal tissue but also in peritoneal macrophages of Nox1-KO. Up-regulation of iNOS by LPS was significantly inhibited in macrophages deficient in Nox1, illustrating a functional hierarchy in NOX1/iNOS signaling. Together, these findings suggest that NOX1 in bone marrow-derived cells, but not epithelial cells, perturbs intestinal barrier integrity during endotoxemia.
Collapse
Affiliation(s)
| | | | - Kai Zhu
- Department of Pharmacology, Japan; Department of Nephrology, Renmin Hospital of Wuhan University, 238 Jiefang Rd., Wuchang District, Wuhan, 430060, China
| | | | - Kenjiro Matsumoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | | | | | | | - Masato Katsuyama
- Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, Okinawa, 903-0215, Japan
| | - Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | | |
Collapse
|
21
|
Quaresma M, Damasceno S, Monteiro C, Lima F, Mendes T, Lima M, Justino P, Barbosa A, Souza M, Souza E, Soares P. Probiotic mixture containing Lactobacillus spp. and Bifidobacterium spp. attenuates 5-fluorouracil-induced intestinal mucositis in mice. Nutr Cancer 2019; 72:1355-1365. [PMID: 31608714 DOI: 10.1080/01635581.2019.1675719] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lactobacillus spp. and Bifidobacterium spp. was used to protect against gastrointestinal disorders. The present study evaluated the effects of probiotic mixture (PM) containing Lactobacillus spp. and Bifidobacterium spp. on intestinal mucositis induced by 5-fluorouracil (5-FU). Swiss male mice (25-30 g) were treated with 5-FU (450 mg/kg, ip) and were orally administered (PM). Probiotic mixture 1 (PM-1) is a mixture of two probiotics (Lactobacillus acidophilus and Bifidobacterium lactis) and probiotic mixture 2 (PM-2) is a mixture of four probiotics (Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus rhamnosus, and Bifidobacterium lactis). PM-1 and PM-2 decreased histopathological scores in the duodenum and jejunum after mucositis. PM-2 attenuated 5-FU-induced weight loss. On the other hand, PM-1 did not exert a significant effect on weight loss. Both probiotics mixture increased the villus/crypt ratio in all intestinal segments, increased GSH levels in the duodenum and jejunum, and reduced the MDA, MPO, TNF-α, and IL-6 levels in the duodenum, jejunum, and ileum. PM-2 attenuated the delay in gastric emptying. PM-1 and PM-2 prevented epithelial injury in intestinal mucositis by 5-FU, demonstrating the potential use of these probiotics as therapeutic agents against intestinal mucositis.
Collapse
Affiliation(s)
- Marielle Quaresma
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Samara Damasceno
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Carlos Monteiro
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Francisco Lima
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Tiago Mendes
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Marcos Lima
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Priscilla Justino
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - André Barbosa
- LAFFEX-Laboratory of Experimental Physiopharmacology, Biotechnology and Biodiversity Center Research, Federal University of Piauí, Parnaíba, Brazil
| | - Marcellus Souza
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil
| | - Emmanuel Souza
- Department of Morphology, Medical School, Federal University of Ceara, Ceara, Brazil
| | - Pedro Soares
- Department of Physiology and Pharmacology, LEFFAG-Laboratory of Physiopharmacology Study of Gastrointestinal Tract, Federal University of Ceará, Fortaleza, Brazil.,Department of Morphology, Medical School, Federal University of Ceara, Ceara, Brazil
| |
Collapse
|
22
|
Wenqin D, Yaodong Z, Wanji S, Fengli Z, Li S, Haili J, Ping L, Mei Z. Armillariella Oral Solution Ameliorates Small Intestinal Damage in a Mouse Model of Chemotherapy-Induced Mucositis. Nutr Cancer 2019; 71:1142-1152. [PMID: 31210536 DOI: 10.1080/01635581.2019.1599029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background: Armillariella oral solution (AOS) shows therapeutic effect on gastrointestinal disorders. We aimed to investigate the potential efficacy of AOS on chemotherapy-induced intestinal mucositis in mice. Methods: Intestinal mucositis was induced in C57BL/6 mice by daily intraperitoneal injection of 5-FU (50 mg/kg) for 7 days. Effects of AOS (at 1, 5, and 10 mL/kg), or combined Bifidobacterium and Lactobacillus (CBL, 450 mg/kg) on the accompanying morphometry and histology, expression of Ki-67, caspase-3, Lgr5 and apoptosis of intestinal crypt cells were assessed. Results: Continuous administration of 5-FU to mice caused severe intestinal mucositis, which was histologically characterized by the destruction of intestinal crypts and shortening of villi, accompanied by diarrhea and body weight loss. Daily AOS administration dose-dependently reduced the severity of intestinal mucositis, diarrhea and body weight loss. Similar beneficial effects were observed with CBL. The expression of Ki-67 and Lgr5 decreased and the expression of caspase-3, and the number of apoptotic cells increased 24 h after the first 5-FU administration (P < 0.05), and these responses were significantly reduced by AOS treatment (P < 0.05, at 5 or 10 mL/kg). Conclusions: AOS can alleviate 5-FU-induced mucositis in mice via increasing Lgr5 expression and suppressing apoptotic responses in the intestinal crypt cells.
Collapse
Affiliation(s)
- Dong Wenqin
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Zhu Yaodong
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Song Wanji
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Zhang Fengli
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Su Li
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Jiang Haili
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Li Ping
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| | - Zhang Mei
- Department of Chinese Integrated Medicine Oncology, First Affiliated Hospital of Anhui Medical University , Hefei , Anhui Province , China
| |
Collapse
|
23
|
Fu C, Chu J, Shen A, Liu L, Chen H, Lin J, Sferra TJ, Chen Y, Peng J. Pien Tze Huang alleviates 5-fluorouracil-induced intestinal mucositis in CT-26 tumor-bearing mice. Exp Ther Med 2017; 14:2291-2297. [PMID: 28962157 PMCID: PMC5609198 DOI: 10.3892/etm.2017.4755] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 03/31/2017] [Indexed: 01/06/2023] Open
Abstract
Chemotherapeutic agents, including 5-fluorouracil (5-FU), frequently cause intestinal mucositis as a side effect, leading to life quality reduction in colorectal cancer (CRC) patients and interruption of CRC treatment. Traditional Chinese medicines (TCMs) have recently received attention due to their relatively few adverse effects. Pien Tze Huang (PZH), a well-known TCM formulation first documented to have been prescribed >450 years ago, has been demonstrated to be clinically effective in treating various types of cancer including CRC. Although previous studies by our group reported that PZH possesses a variety of anti-cancer activities via multiple mechanisms, it has remained elusive whether it is able to reduce intestinal mucositis induced by 5-FU. The present study evaluated the effect of PZH on 5-FU-induced intestinal mucositis in CT-26 tumor-bearing xenograft mice and investigated the possible molecular mechanism. The results indicated that administration of PZH effectively alleviated the severity of 5-FU-induced diarrhea and morphological intestinal damages, but had no significant effect on body weight loss. In addition, a terminal deoxynucleotidyl transferase dUTP nick end labeling assay revealed that PZH treatment significantly inhibited cell apoptosis in the intestinal crypt. Furthermore, immunohistochemical staining showed that PZH treatment reduced the protein expression of pro-apoptotic B-cell lymphoma 2 (Bcl-2)-associated X protein but enhanced that of anti-apoptotic Bcl-2 in the intestinal crypt. Taken together, the results of the present study suggested that PZH effectively attenuates 5-FU-induced intestinal mucositis, which is in part associated with its inhibitory effect on cell apoptosis in the intestinal crypt.
Collapse
Affiliation(s)
- Caixuan Fu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Pediatric Department, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Hongwei Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Thomas J. Sferra
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Pediatric Department, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Youqin Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Pediatric Department, Rainbow Babies and Children's Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
24
|
Kato S, Hamouda N, Kano Y, Oikawa Y, Tanaka Y, Matsumoto K, Amagase K, Shimakawa M. Probiotic Bifidobacterium bifidum
G9-1 attenuates 5-fluorouracil-induced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses. Clin Exp Pharmacol Physiol 2017; 44:1017-1025. [DOI: 10.1111/1440-1681.12792] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/30/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Shinichi Kato
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Nahla Hamouda
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Yoshitaro Kano
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Yousuke Oikawa
- R&D Center; Biofermin Pharmaceutical Co., Ltd.; Kobe Japan
| | - Yoshiki Tanaka
- R&D Center; Biofermin Pharmaceutical Co., Ltd.; Kobe Japan
| | - Kenjiro Matsumoto
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | - Kikuko Amagase
- Division of Pathological Sciences; Department of Pharmacology and Experimental Therapeutics; Kyoto Pharmaceutical University; Kyoto Japan
| | | |
Collapse
|
25
|
Aviello G, Knaus UG. ROS in gastrointestinal inflammation: Rescue Or Sabotage? Br J Pharmacol 2017; 174:1704-1718. [PMID: 26758851 PMCID: PMC5446568 DOI: 10.1111/bph.13428] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/09/2015] [Accepted: 01/07/2016] [Indexed: 12/15/2022] Open
Abstract
The intestine is composed of many distinct cell types that respond to commensal microbiota or pathogens with immune tolerance and proinflammatory signals respectively. ROS produced by mucosa-resident cells or by newly recruited innate immune cells are essential for antimicrobial responses and regulation of signalling pathways including processes involved in wound healing. Impaired ROS production due to inactivating patient variants in genes encoding NADPH oxidases as ROS source has been associated with Crohn's disease and pancolitis, whereas overproduction of ROS due to up-regulation of oxidases or altered mitochondrial function was linked to ileitis and ulcerative colitis. Here, we discuss recent advances in our understanding of how maintaining a redox balance is crucial to preserve gut homeostasis. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- G Aviello
- National Children's Research CentreOur Lady's Children's HospitalDublinIreland
| | - UG Knaus
- National Children's Research CentreOur Lady's Children's HospitalDublinIreland
- Conway Institute, School of MedicineUniversity College DublinDublinIreland
| |
Collapse
|
26
|
Hamouda N, Sano T, Oikawa Y, Ozaki T, Shimakawa M, Matsumoto K, Amagase K, Higuchi K, Kato S. Apoptosis, Dysbiosis and Expression of Inflammatory Cytokines are Sequential Events in the Development of 5-Fluorouracil-Induced Intestinal Mucositis in Mice. Basic Clin Pharmacol Toxicol 2017; 121:159-168. [PMID: 28374966 DOI: 10.1111/bcpt.12793] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/29/2017] [Indexed: 12/26/2022]
Abstract
The chemotherapeutic agent 5-fluorouracil (5-FU) causes intestinal mucositis with severe diarrhoea, but the pathogenesis is not fully understood. In this study, we investigated the pathogenic effects of 5-FU in mice, focusing on apoptosis, enterobacteria and inflammatory cytokines. Repeated administration of 5-FU caused severe intestinal mucositis on day 6, accompanied by diarrhoea and body-weight loss. TNF-α expression increased 1 day after exposure to the drug, and spiked a second time on day 4, at which point myeloperoxidase activity and IL-1β expression also increased. Apoptotic cells were observed in intestinal crypts only on day 1. 5-FU also induced dysbiosis, notably decreasing the abundance of intestinal Firmicutes while increasing the abundance of Bacteroidetes and Verrucomicrobia. Twice-daily co-administration of oral antibiotics significantly reduced the severity of intestinal mucositis and dysbiosis, and blocked the increase in myeloperoxidase activity and cytokine expression on day 6, without affecting apoptosis and TNF-α up-regulation on day 1. In cultured colonic epithelial cells, exposure to 5-FU also up-regulated TNF-α expression. Collectively, the data suggest that crypt apoptosis, dysbiosis and expression of inflammatory cytokines are sequential events in the development of intestinal mucositis after exposure to 5-FU. In particular, 5-FU appears to directly induce apoptosis via TNF-α and to suppress intestinal cell proliferation, thereby resulting in degradation of the epithelial barrier, as well as in secondary inflammation mediated by inflammatory cytokines.
Collapse
Affiliation(s)
- Nahla Hamouda
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Tatsushi Sano
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan.,Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Yosuke Oikawa
- R&D Center, Biofermin Pharmaceutical Co., Ltd., Kobe, Hyogo, Japan
| | - Toru Ozaki
- R&D Center, Biofermin Pharmaceutical Co., Ltd., Kobe, Hyogo, Japan
| | - Masaki Shimakawa
- R&D Center, Biofermin Pharmaceutical Co., Ltd., Kobe, Hyogo, Japan
| | - Kenjiro Matsumoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kikuko Amagase
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Kazuhide Higuchi
- Second Department of Internal Medicine, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
27
|
Abalo R, Uranga JA, Pérez-García I, de Andrés R, Girón R, Vera G, López-Pérez AE, Martín-Fontelles MI. May cannabinoids prevent the development of chemotherapy-induced diarrhea and intestinal mucositis? Experimental study in the rat. Neurogastroenterol Motil 2017; 29. [PMID: 27686064 DOI: 10.1111/nmo.12952] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND The antineoplastic drug 5-fluoruracil (5-FU) is a pirimidine analog, which frequently induces potentially fatal diarrhea and mucositis. Cannabinoids reduce gastrointestinal motility and secretion and might prevent 5-FU-induced gut adverse effects. Here, we asked whether cannabinoids may prevent diarrhea and mucositis induced by 5-FU in the rat. METHODS Male Wistar rats received vehicle or the non-selective cannabinoid agonist WIN 55,212-2 (WIN; 0.5 mg kg-1 injection-1 , 1 injection day-1 , 4 consecutive days) by intraperitoneal (ip) route; on the first 2 days, animals received also saline or 5-FU (150 mg kg-1 injection-1 , cumulative dose of 300 mg kg-1 ). Gastrointestinal motor function was radiographically studied after barium contrast intragastric administration on experimental days 1 and 4. Structural alterations of the stomach, small intestine and colon were histologically studied on day 4. PAS staining and immunohistochemistry for Ki67, chromogranin A and CD163 were used to detect secretory, proliferating, and endocrine cells, and activated macrophages respectively. KEY RESULTS As shown radiographically, 5-FU induced significant gastric emptying delay (on days 1 and 4) and diarrhea (on day 4). WIN did not significantly alter the motility curves obtained for either control or 5-FU-treated animals but tended to reduce the severity of 5-FU-induced diarrhea and increased permanence of barium from day 1 to the beginning of day 4 in 5-FU-treated animals. 5-FU-induced mucositis was severe and not counteracted by WIN. CONCLUSIONS AND INFERENCES 5-FU-induced diarrhea, but not mucositis, was partly prevented by WIN at a low dose. Cannabinoids might be useful to prevent chemotherapy-induced diarrhea.
Collapse
Affiliation(s)
- R Abalo
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain
| | - J A Uranga
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain
| | - I Pérez-García
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain
| | - R de Andrés
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain
| | - R Girón
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain
| | - G Vera
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain
| | - A E López-Pérez
- Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain.,Unidad del Dolor, Servicio de Anestesiología, Hospital General Universitario Gregorio Marañón (HGUGM), Madrid, Spain
| | - M I Martín-Fontelles
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Madrid, Alcorcón, Spain.,Unidad Asociada I+D+i al Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC), Madrid, Spain.,Unidad Asociada I+D+i al Instituto de Química Médica, IQM (CSIC), Madrid, Spain.,Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Madrid, Spain
| |
Collapse
|
28
|
Koizumi R, Azuma K, Izawa H, Morimoto M, Ochi K, Tsuka T, Imagawa T, Osaki T, Ito N, Okamoto Y, Saimoto H, Ifuku S. Oral Administration of Surface-Deacetylated Chitin Nanofibers and Chitosan Inhibit 5-Fluorouracil-Induced Intestinal Mucositis in Mice. Int J Mol Sci 2017; 18:ijms18020279. [PMID: 28134832 PMCID: PMC5343815 DOI: 10.3390/ijms18020279] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 01/24/2017] [Indexed: 01/27/2023] Open
Abstract
This study investigated the prophylactic effects of orally administered surface-deacetylated chitin nanofibers (SDACNFs) and chitosan against 5-fluorouracil (5-FU)-induced intestinal mucositis, which is a common side effect of 5-FU chemotherapy. SDACNFs and chitosan abolished histological abnormalities associated with intestinal mucositis and suppressed hypoproliferation and apoptosis of intestinal crypt cells. These results indicate that SDACNF and chitosan are useful agents for preventing mucositis induced by anti-cancer drugs.
Collapse
Affiliation(s)
- Ryo Koizumi
- Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan.
| | - Kazuo Azuma
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Hironori Izawa
- Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan.
| | - Minoru Morimoto
- Division of Instrumental Analysis, Research Center for Bioscience and Technology, Tottori University, Tottori 680-8550, Japan.
| | - Kosuke Ochi
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Takeshi Tsuka
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Tomohiro Imagawa
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Tomohiro Osaki
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Norihiko Ito
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Yoshiharu Okamoto
- Department of Veterinary Clinical Medicine, Tottori University, Tottori 680-8553, Japan.
| | - Hiroyuki Saimoto
- Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan.
| | - Shinsuke Ifuku
- Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan.
| |
Collapse
|
29
|
Kuchay RAH. A review of complementary therapies for chemotherapy induced gastrointestinal mucositis. Drug Discov Ther 2017; 10:292-299. [PMID: 27746417 DOI: 10.5582/ddt.2016.01059] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Administration of chemotherapy often leads to gastrointestinal mucositis (GIM). GIM manifests as nausea, abdominal pain and diarrhoea in recipients of chemotherapy. GIM is a major complication occurring in approximately 80% of patients receiving 5-flurouracil treatment. These side-effects may become so severe that significant dose reductions are required, ultimately affecting treatment efficacy and patient survival. Complementary and alternative medicine (CAM) is a growing area of public interest. This review will provide an overview of current knowledge of complementary medicinal therapies for chemotherapy induced GIM. An understanding of this evolving literature is useful in discussing these therapies with patients who are considering using them.
Collapse
|
30
|
Yokota H, Tsuzuki A, Shimada Y, Imai A, Utsumi D, Tsukahara T, Matsumoto M, Amagase K, Iwata K, Nakamura A, Yabe-Nishimura C, Kato S. NOX1/NADPH Oxidase Expressed in Colonic Macrophages Contributes to the Pathogenesis of Colonic Inflammation in Trinitrobenzene Sulfonic Acid-Induced Murine Colitis. J Pharmacol Exp Ther 2017; 360:192-200. [PMID: 27754929 DOI: 10.1124/jpet.116.235580] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 10/14/2016] [Indexed: 03/08/2025] Open
Abstract
NOX1/NADPH oxidase, a nonphagocytic isoform of reactive oxygen species-producing enzymes, is highly expressed in the colon, but the physiologic and pathophysiologic roles of this isoform are not fully understood. The present study investigated the role of NOX1 in the development of colonic inflammation in a trinitrobenzene sulfonic acid (TNBS)-induced murine colitis model. Intrarectal injection of TNBS caused severe colitis accompanied by body weight loss, diarrhea, and increased myeloperoxidase (MPO) activity in wild-type (WT) mice. In contrast, the severity of colitis was significantly attenuated in NOX1-deficient (NOX1KO) mice (the inhibitions of macroscopic damage score, body weight loss, diarrhea score, and MPO activity were 73.1%, 36.8%, 83.3%, and 98.4%, respectively). TNBS-induced upregulation of inflammatory cytokines (tumor necrosis factor (TNF)-α and interleukin (IL)-1β), chemokines (CXCL1 and CXLC2), and inducible nitric oxide synthase (iNOS) was also significantly less in NOX1KO than in WT mice (the inhibitions were 100.8%, 89.0%, 63.5%, 96.7%, and 97.1%, respectively). Expression of NOX1 mRNA was detected not only in the lamina propria but also in peritoneal macrophages isolated from WT mice. Increased expression of TNF-α, IL-1β, and iNOS in peritoneal macrophages exposed to lipopolysaccharide was significantly attenuated in macrophages isolated from NOX1KO mice (68.1%, 67.0%, and 79.3% inhibition, respectively). These findings suggest that NOX1/NADPH oxidase plays an important role in the pathogenesis of TNBS-induced colonic inflammation via upregulation of inflammatory cytokines, chemokines, and iNOS. NOX1 in colonic macrophages may become a potential target in pharmacologic intervention for inflammatory bowel disease.
Collapse
Affiliation(s)
- Haruka Yokota
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Ayaka Tsuzuki
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Yuki Shimada
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Azusa Imai
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Daichi Utsumi
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Takuya Tsukahara
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Misaki Matsumoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Kikuko Amagase
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Kazumi Iwata
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Akio Nakamura
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Chihiro Yabe-Nishimura
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| | - Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University (H.Y., A.T., Y. S., A. I., D. U., T. T., K. A., S.K.), and Department of Pharmacology, Kyoto Prefectural University of Medicine (M.M., K.I., C.Y-N.), Kyoto, Japan, and Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan (A.N.)
| |
Collapse
|
31
|
Sakai H, Tabata S, Kimura M, Yabe S, Isa Y, Kai Y, Sato F, Yumoto T, Miyano K, Narita M, Uezono Y. Active Ingredients of Hange-shashin-to, Baicalelin and 6-Gingerol, Inhibit 5-Fluorouracil-Induced Upregulation of CXCL1 in the Colon to Attenuate Diarrhea Development. Biol Pharm Bull 2017; 40:2134-2139. [DOI: 10.1248/bpb.b17-00479] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hiroyasu Sakai
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Shoko Tabata
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Minami Kimura
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Saori Yabe
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Yosuke Isa
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Yuki Kai
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Fumiaki Sato
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Tetsuro Yumoto
- Department of Analytical Pathophysiology, Division of Pharmacy Professional Development and Research, Hoshi University
| | - Kanako Miyano
- Division of Cancer Pathophysiology, National Cancer Center Research Institute
| | | | - Yasuhito Uezono
- Division of Cancer Pathophysiology, National Cancer Center Research Institute
- Division of Supportive Cancer Research, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center
- Innovation Center for Supportive, Palliative and Psychosocial Care, National Cancer Center Hospital
| |
Collapse
|
32
|
Seo K, Ki SH, Park EY, Shin SM. 5-Fluorouracil inhibits cell migration by induction of Sestrin2 in colon cancer cells. Arch Pharm Res 2016; 40:231-239. [PMID: 28028695 DOI: 10.1007/s12272-016-0878-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/17/2016] [Indexed: 01/04/2023]
Abstract
5-Fluorouracil (5-FU) is a chemotherapeutic agent used in the treatment of colorectal cancer. In this study, we investigated whether 5-FU induces Sestrin2 (SESN2), an antioxidant enzyme, and the role of SESN2 in 5-FU action in colon cancer cells. We found that 5-FU upregulated SESN2 protein expression in both HCT116 and HT29 cells. It also increased transcripts of SESN1 and SESN2, but not of SESN3. Furthermore, we investigated whether production of reactive oxygen species (ROS) was involved in 5-FU-induced SESN2 expression. 5-FU did not increase ROS production nor affect Nrf2 phosphorylation and expression levels. Moreover, SESN2 upregulation by 5-FU was not prevented by pretreatment with antioxidants. Next, we investigated p53 levels after 5-FU treatment to elucidate the regulation of SESN2 by 5-FU. An increase in p53 levels was detected following 5-FU treatment; pifithrin-α, an inhibitor of p53 activation, reversed 5-FU-induced SESN2 expression. 5-FU prevented serum-induced in vitro cell migration, but knockdown of SESN2 or treatment with pifithrin-α reversed a 5-FU-mediated decrease in cell migration. Taken together, our results suggest that 5-FU increases SESN2 levels via a p53-dependent pathway, which contributes to inhibition of cancer cell migration in vitro.
Collapse
Affiliation(s)
- Kyuhwa Seo
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, South Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, South Korea
| | - Eun Young Park
- College of Pharmacy, Mokpo National University, Muan-gun, Jeonnam, 534-729, South Korea
| | - Sang Mi Shin
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, South Korea.
| |
Collapse
|
33
|
Mashtoub S, Lampton LS, Eden GL, Cheah KY, Lymn KA, Bajic JE, Howarth GS. Emu Oil Combined with Lyprinol™ Reduces Small Intestinal Damage in a Rat Model of Chemotherapy-Induced Mucositis. Nutr Cancer 2016; 68:1171-1180. [PMID: 27618153 DOI: 10.1080/01635581.2016.1208829] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chemotherapy-induced mucositis is characterized by inflammation and ulcerating lesions lining the alimentary tract. Emu Oil and Lyprinol™ have independently demonstrated their therapeutic potential in intestinal inflammatory disorders, including mucositis. We investigated Emu Oil and Lyprinol™ in combination for their further potential to alleviate chemotherapy-induced mucositis in rats. Rats were gavaged with (1 ml) water, Olive Oil, Emu Oil + Olive Oil, Lyprinol™ + Olive Oil or Emu Oil + Lyprinol™ from Days 0 to 7, injected with saline (control) or 5-Fluorouracil (5-FU) on Day 5 and euthanized on Day 8. Myeloperoxidase (MPO) activity (indicative of acute inflammation), histological severity scores, and intestinal architecture were quantified. Myeloperoxidase activity was significantly increased in the jejunum and ileum following 5-FU, compared to saline controls. Both Olive Oil and Emu Oil + Lyprinol™ significantly reduced jejunal MPO levels (1.8-fold and 1.7-fold, respectively), whereas only Emu Oil + Lyprinol™ significantly decreased ileal MPO levels, relative to 5-FU controls. All oil treatments decreased histological severity scores in the jejunum and ileum, and normalized crypt depth in the mid small intestine, relative to 5-FU controls. Emu Oil combined with Lyprinol™ partially reduced acute small intestinal inflammation. Isolating bioactive constituents of these naturally sourced oils could provide a more targeted strategy to protect against intestinal mucositis.
Collapse
Affiliation(s)
- Suzanne Mashtoub
- a School of Medicine and Pharmacology, The University of Western Australia, Harry Perkins Institute for Medical Research, Fiona Stanley Hospital , Murdoch , Western Australia
- b Department of Gastroenterology , Women's and Children's Hospital , North Adelaide , South Australia
- c Discipline of Physiology, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide , Adelaide , South Australia
| | - Lorrinne S Lampton
- d School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus , Roseworthy , South Australia
| | - Georgina L Eden
- d School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus , Roseworthy , South Australia
| | - Ker Y Cheah
- b Department of Gastroenterology , Women's and Children's Hospital , North Adelaide , South Australia
| | - Kerry A Lymn
- d School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus , Roseworthy , South Australia
- e Women's and Children's Health Research Institute, Women's and Children's Hospital , North Adelaide , South Australia
| | - Juliana E Bajic
- c Discipline of Physiology, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide , Adelaide , South Australia
| | - Gordon S Howarth
- b Department of Gastroenterology , Women's and Children's Hospital , North Adelaide , South Australia
- c Discipline of Physiology, School of Medical Sciences, Faculty of Health Sciences, The University of Adelaide , Adelaide , South Australia
- d School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy Campus , Roseworthy , South Australia
| |
Collapse
|
34
|
Irinotecan- and 5-fluorouracil-induced intestinal mucositis: insights into pathogenesis and therapeutic perspectives. Cancer Chemother Pharmacol 2016; 78:881-893. [PMID: 27590709 DOI: 10.1007/s00280-016-3139-y] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/23/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE Intestinal mucositis and diarrhea are common manifestations of anticancer regimens that include irinotecan, 5-fluorouracil (5-FU), and other cytotoxic drugs. These side effects negatively impact therapeutic outcomes and delay subsequent cycles of chemotherapy, resulting in dose reductions and treatment discontinuation. Here, we aimed to review the experimental evidence regarding possible new targets for the management of irinotecan- and 5-FU-related intestinal mucositis. METHODS A literature search was performed using the PubMed and MEDLINE databases. No publication time limit was set for article inclusion. RESULTS Here, we found that clinical management of intestinal mucositis and diarrhea is somewhat ineffective at reducing symptoms, possibly due to a lack of specific targets for modulation. We observed that IL-1β contributes to the apoptosis of enterocytes in mucositis induced by 5-FU. However, 5-FU-related mucositis is far less thoroughly investigated with regard to specific molecular targets when compared to irinotecan-related disease. Several studies have proposed that a correlation exists between the intestinal microbiota, the enterohepatic recirculation of active metabolites of irinotecan, and the establishment of mucositis. However, as reviewed here, this association seems to be controversial. In addition, the pathogenesis of irinotecan-induced mucositis appears to be orchestrated by interleukin-1/Toll-like receptor family members, leading to epithelial cell apoptosis. CONCLUSIONS IL-1β, IL-18, and IL-33 and the receptors IL-1R, IL-18R, ST2, and TLR-2 are potential therapeutic targets that can be modulated to minimize anticancer agent-associated toxicity, optimize cancer treatment dosing, and improve clinical outcomes. In this context, the pathogenesis of mucositis caused by other anticancer agents should be further investigated.
Collapse
|
35
|
Kwon Y. Mechanism-based management for mucositis: option for treating side effects without compromising the efficacy of cancer therapy. Onco Targets Ther 2016; 9:2007-16. [PMID: 27103826 PMCID: PMC4827894 DOI: 10.2147/ott.s96899] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mucositis is a major side effect induced by chemotherapy and radiotherapy. Although mucositis is a leading cause of morbidity and mortality in cancer patients, management is largely limited to controlling symptoms, and few therapeutic agents are available for treatment. Since mucositis could be inhibited by the modulation of radiotherapy- or chemotherapy-induced pathways independently of cancer treatment, there is an opportunity for the development of more targeted therapies and interventions. This article examined potential therapeutic agents that have been investigated for the prevention and/or inhibition of mucositis induced by conventional chemotherapy and radiotherapy. They can be classified according to their mechanisms of action: scavenging reactive oxygen species, inhibition of specific cytokine production or inflammation, and inhibition of apoptosis. These early events may be good target pathways for preventing the pathogenesis of mucositis. Considering that both cancer therapy and therapeutic agents for mucositis act on both normal and cancer cells, agents that inhibit mucositis should act through mechanisms that selectively protect normal cells without compromising cancer treatment. Therefore, mechanism-based guidance for the treatment of mucositis is critical to prevent risky treatments for cancer patients and to relieve detrimental side effects effectively from cancer therapy.
Collapse
Affiliation(s)
- Youngjoo Kwon
- Department of Food Science and Engineering, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
36
|
Deliyanti D, Wilkinson-Berka JL. Inhibition of NOX1/4 with GKT137831: a potential novel treatment to attenuate neuroglial cell inflammation in the retina. J Neuroinflammation 2015. [PMID: 26219952 PMCID: PMC4518508 DOI: 10.1186/s12974-015-0363-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inflammation and the excess production of reactive oxygen species (ROS) contribute significantly to the pathogenesis of ischemic retinopathies such as diabetic retinopathy and retinopathy of prematurity. We hypothesized that GKT137831, a dual inhibitor of NADPH oxidases (NOX) 1 and NOX4, reduces inflammation in the ischemic retina by dampening the pro-inflammatory phenotype of retinal immune cells as well as macroglial Müller cells and neurons. METHODS Ischemic retinopathy was induced in Sprague-Dawley rats by exposure to 80 % O2 cycled with 21 % O2 for 3 h per day from postnatal day (P) 0 to P11, followed by room air (P12 to P18). GKT137831 was administered P12 to P18 (60 mg/kg, subcutaneous) and comparisons were to room air controls. Retinal inflammation was examined by measuring leukocyte adherence to the retinal vasculature, ionized calcium-binding adaptor protein-1-positive microglia/macrophages, and the mRNA and protein levels of key inflammatory factors involved in retinal disease. Damage to Müller cells was evaluated by quantitating glial fibrillary acidic protein-positive cells and vascular leakage with an albumin ELISA. To verify the anti-inflammatory actions of GKT137831 on glia and neurons involved in ischemic retinopathy, primary cultures of rat retinal microglia, Müller cells, and ganglion cells were exposed to the in vitro counterpart of ischemia, hypoxia (0.5 %), and treated with GKT137831 for up to 72 h. ROS levels were evaluated with dihydroethidium and the protein and gene expression of inflammatory factors with quantitative PCR, ELISA, and a protein cytokine array. RESULTS In the ischemic retina, GKT137831 reduced the increased leukocyte adherence to the vasculature, the pro-inflammatory phenotype of microglia and macroglia, the increased gene and protein expression of vascular endothelial growth factor, monocyte chemoattractant protein-1, and leukocyte adhesion molecules as well as vascular leakage. In all cultured cell types, GKT137831 reduced the hypoxia-induced increase in ROS levels and protein expression of various inflammatory mediators. CONCLUSIONS NOX1/4 enzyme inhibition with GKT137831 has potent anti-inflammatory effects in the retina, indicating its potential as a treatment for a variety of vision-threatening retinopathies.
Collapse
Affiliation(s)
- Devy Deliyanti
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct Level 6, 89 Commercial Road, Melbourne, VIC, Australia, 3004
| | - Jennifer L Wilkinson-Berka
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct Level 6, 89 Commercial Road, Melbourne, VIC, Australia, 3004.
| |
Collapse
|
37
|
Kojouharov BM, Brackett CM, Veith JM, Johnson CP, Gitlin II, Toshkov IA, Gleiberman AS, Gudkov AV, Burdelya LG. Toll-like receptor-5 agonist Entolimod broadens the therapeutic window of 5-fluorouracil by reducing its toxicity to normal tissues in mice. Oncotarget 2015; 5:802-14. [PMID: 24583651 PMCID: PMC3996654 DOI: 10.18632/oncotarget.1773] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Myelosuppression and gastrointestinal damage are common side effects of cancer treatment limiting efficacy of DNA-damaging chemotherapeutic drugs. The Toll-like receptor 5 (TLR5) agonist Entolimod has demonstrated efficacy in mitigating damage to hematopoietic and gastrointestinal tissues caused by radiation. Here, using 5-Fluorouracil (5-FU) treated mice as a model of chemotherapy-induced side effects, we demonstrated significant reduction in the severity of 5-FU-induced morbidity and increased survival accompanied by the improved integrity of intestinal tissue and stimulated the restoration of hematopoiesis. Entolimod-stimulated IL-6 production was essential for Entolimod's ability to rescue mice from death caused by doses of 5-FU associated with hematopoietic failure. In contrast, IL-6 induction was not necessary for protection and restoration of drug-damaged gastrointestinal tissue by Entolimod. In a syngeneic mouse CT26 colon adenocarcinoma model, Entolimod reduced the systemic toxicity of 5-FU, but did not reduce its antitumor efficacy indicating that the protective effect of Entolimod was selective for normal, non-tumor, tissues. These results suggest that Entolimod has clinical potential to broaden the therapeutic window of genotoxic anticancer drugs by reducing their associated hematopoietic and gastrointestinal toxicities.
Collapse
|
38
|
Kato S, Hayashi S, Kitahara Y, Nagasawa K, Aono H, Shibata J, Utsumi D, Amagase K, Kadowaki M. Saireito (TJ-114), a Japanese traditional herbal medicine, reduces 5-fluorouracil-induced intestinal mucositis in mice by inhibiting cytokine-mediated apoptosis in intestinal crypt cells. PLoS One 2015; 10:e0116213. [PMID: 25565296 PMCID: PMC4286213 DOI: 10.1371/journal.pone.0116213] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 12/05/2014] [Indexed: 12/30/2022] Open
Abstract
Clinical chemotherapy frequently causes intestinal mucositis as a side effect, which is accompanied by severe diarrhea. We recently showed that the cytokine-mediated apoptotic pathway might be important for the development of intestinal mucositis induced by 5-fluorouracil (5-FU). Saireito, the traditional Japanese herbal (Kampo) medicine, is widely used to treat diarrhea and various inflammatory diseases in Japan. In the present study, we investigated the effect of saireito on 5-FU-induced intestinal mucositis in mice, especially in relation to apoptosis in the intestinal crypt. Male C57BL/6 mice were given 5-FU (50 mg/kg), i.p. once daily for 6 days. Intestinal mucositis was evaluated histochemically. Saireito (100–1000 mg/kg) was administered p.o. twice daily for 6 days. Repeated 5-FU treatment caused severe intestinal mucositis including morphological damage, which was accompanied by body weight loss and diarrhea. Daily administration of saireito reduced the severity of intestinal mucositis in a dose-dependent manner. Body weight loss and diarrhea during 5-FU treatment were also significantly attenuated by saireito administration. The number of apoptotic and caspase-3-activated cells in the intestinal crypt was increased, and was accompanied by up-regulated tumor necrosis factor (TNF)-α and interleukin (IL)-1β mRNA within 24 h of the first 5-FU injection. However, all of these measures were significantly lower after saireito administration. These results suggest that saireito attenuates 5-FU-induced intestinal mucositis. This action may come from the reduction of apoptosis in the intestinal crypt via suppression of the up-regulation of inflammatory cytokines. Therefore, saireito may be clinically useful for the prevention of intestinal mucositis during cancer chemotherapy.
Collapse
Affiliation(s)
- Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
- * E-mail:
| | - Shusaku Hayashi
- Division of Gastrointestinal Pathophysiology, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Sugitani, Toyama 930–0194, Japan
| | - Yumeno Kitahara
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
| | - Koyo Nagasawa
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
| | - Hitomi Aono
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
| | - Junichiro Shibata
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
| | - Daichi Utsumi
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
| | - Kikuko Amagase
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Misasagi, Yamashina, Kyoto 607–8414, Japan
| | - Makoto Kadowaki
- Division of Gastrointestinal Pathophysiology, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Sugitani, Toyama 930–0194, Japan
| |
Collapse
|
39
|
Welak SR, Rentea RM, Teng RJ, Heinzerling N, Biesterveld B, Liedel JL, Pritchard KA, Fredrich KM, Gourlay DM. Intestinal NADPH oxidase 2 activity increases in a neonatal rat model of necrotizing enterocolitis. PLoS One 2014; 9:e115317. [PMID: 25517730 PMCID: PMC4269454 DOI: 10.1371/journal.pone.0115317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/21/2014] [Indexed: 12/21/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a complication of prematurity. The etiology is unknown, but is related to enteral feeding, ischemia, infection, and inflammation. Reactive oxygen species production, most notably superoxide, increases in NEC. NADPH oxidase (NOX) generates superoxide, but its activity in NEC remains unknown. We hypothesize that NOX-derived superoxide production increases in NEC. Newborn Sprague-Dawley rats were divided into control, formula-fed, formula/LPS, formula/hypoxia, and NEC (formula, hypoxia, and LPS). Intestinal homogenates were analyzed for NADPH-dependent superoxide production. Changes in superoxide levels on days 0-4 were measured. Inhibitors for nitric oxide synthase (L-NAME) and NOX2 (GP91-ds-tat) were utilized. RT-PCR for eNOS, NOX1, GP91phox expression was performed. Immunofluorescence studies estimated the co-localization of p47phox and GP91phox in control and NEC animals on D1, D2, and D4. NEC pups generated more superoxide than controls on D4, while all other groups were unchanged. NADPH-dependent superoxide production was greater in NEC on days 0, 3, and 4. GP91-ds-tat decreased superoxide production in both groups, with greater inhibition in NEC. L-NAME did not alter superoxide production. Temporally, superoxide production varied minimally in controls. In NEC, superoxide generation was decreased on day 1, but increased on days 3-4. GP91phox expression was higher in NEC on days 2 and 4. NOX1 and eNOS expression were unchanged from controls. GP91phox and p47phox had minimal co-localization in all control samples and NEC samples on D1 and D2, but had increased co-localization on D4. In conclusion, this study proves that experimentally-induced NEC increases small intestinal NOX activity. All components of NEC model are necessary for increased NOX activity. NOX2 is the major source, especially as the disease progresses.
Collapse
Affiliation(s)
- Scott R. Welak
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Translational Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Rebecca M. Rentea
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ru-Jeng Teng
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Translational Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Nathan Heinzerling
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Ben Biesterveld
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Jennifer L. Liedel
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Critical Care, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Kirkwood A. Pritchard
- Translational Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Katherine M. Fredrich
- Translational Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - David M. Gourlay
- Children's Research Institute, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Translational Vascular Biology Program, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
40
|
Arifa RD, Madeira MF, de Paula TP, Lima RL, Tavares LD, Menezes-Garcia Z, Fagundes CT, Rachid MA, Ryffel B, Zamboni DS, Teixeira MM, Souza DG. Inflammasome Activation Is Reactive Oxygen Species Dependent and Mediates Irinotecan-Induced Mucositis through IL-1β and IL-18 in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2023-34. [DOI: 10.1016/j.ajpath.2014.03.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 02/28/2014] [Accepted: 03/11/2014] [Indexed: 01/29/2023]
|
41
|
Schwab L, Goroncy L, Palaniyandi S, Gautam S, Triantafyllopoulou A, Mocsai A, Reichardt W, Karlsson FJ, Radhakrishnan SV, Hanke K, Schmitt-Graeff A, Freudenberg M, von Loewenich FD, Wolf P, Leonhardt F, Baxan N, Pfeifer D, Schmah O, Schönle A, Martin SF, Mertelsmann R, Duyster J, Finke J, Prinz M, Henneke P, Häcker H, Hildebrandt GC, Häcker G, Zeiser R. Neutrophil granulocytes recruited upon translocation of intestinal bacteria enhance graft-versus-host disease via tissue damage. Nat Med 2014; 20:648-54. [PMID: 24836575 DOI: 10.1038/nm.3517] [Citation(s) in RCA: 222] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 03/03/2014] [Indexed: 12/15/2022]
Abstract
Acute graft-versus-host disease (GVHD) considerably limits wider usage of allogeneic hematopoietic cell transplantation (allo-HCT). Antigen-presenting cells and T cells are populations customarily associated with GVHD pathogenesis. Of note, neutrophils are the largest human white blood cell population. The cells cleave chemokines and produce reactive oxygen species, thereby promoting T cell activation. Therefore, during an allogeneic immune response, neutrophils could amplify tissue damage caused by conditioning regimens. We analyzed neutrophil infiltration of the mouse ileum after allo-HCT by in vivo myeloperoxidase imaging and found that infiltration levels were dependent on the local microbial flora and were not detectable under germ-free conditions. Physical or genetic depletion of neutrophils reduced GVHD-related mortality. The contribution of neutrophils to GVHD severity required reactive oxygen species (ROS) because selective Cybb (encoding cytochrome b-245, beta polypeptide, also known as NOX2) deficiency in neutrophils impairing ROS production led to lower levels of tissue damage, GVHD-related mortality and effector phenotype T cells. Enhanced survival of Bcl-xL transgenic neutrophils increased GVHD severity. In contrast, when we transferred neutrophils lacking Toll-like receptor-2 (TLR2), TLR3, TLR4, TLR7 and TLR9, which are normally less strongly activated by translocating bacteria, into wild-type C57BL/6 mice, GVHD severity was reduced. In humans, severity of intestinal GVHD strongly correlated with levels of neutrophils present in GVHD lesions. This study describes a new potential role for neutrophils in the pathogenesis of GVHD in both mice and humans.
Collapse
Affiliation(s)
- Lukas Schwab
- 1] Department of Hematology and Oncology, University Medical Center, Freiburg, Germany. [2]
| | - Luise Goroncy
- 1] Department of Hematology and Oncology, University Medical Center, Freiburg, Germany. [2] Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany. [3]
| | - Senthilnathan Palaniyandi
- 1] Division of Hematology and Oncology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA. [2] Division of Bone Marrow Transplantation, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah, USA. [3]
| | - Sanjivan Gautam
- 1] Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany. [2] Department of Medical Microbiology and Hygiene, University Medical Center, Freiburg, Germany. [3] Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University Freiburg, Germany
| | | | - Attila Mocsai
- Department of Physiology, Semmelweis University School of Medicine, Budapest, Hungary
| | - Wilfried Reichardt
- Department of Radiology Medical Physics, University Medical Center, Freiburg, Germany
| | - Fridrik J Karlsson
- Division of Hematology and Oncology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| | - Sabarinath V Radhakrishnan
- 1] Division of Hematology and Oncology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA. [2] Division of Bone Marrow Transplantation, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Kathrin Hanke
- 1] Department of Hematology and Oncology, University Medical Center, Freiburg, Germany. [2] Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Annette Schmitt-Graeff
- Department of Pathology, University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - Marina Freudenberg
- 1] Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany. [2] Centre for Biological Signaling Studies BIOSS, Albert-Ludwigs-University Freiburg, Germany
| | | | - Philipp Wolf
- Department of Urology, University Medical Center, Freiburg, Germany
| | - Franziska Leonhardt
- 1] Department of Hematology and Oncology, University Medical Center, Freiburg, Germany. [2] Faculty of Biology, Albert-Ludwigs-University, Freiburg, Germany
| | - Nicoleta Baxan
- Department of Radiology Medical Physics, University Medical Center, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Oliver Schmah
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Anne Schönle
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Stefan F Martin
- Allergy Research Group, Department of Dermatology, University Medical Center, University Freiburg, Germany
| | - Roland Mertelsmann
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Justus Duyster
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Jürgen Finke
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany
| | - Marco Prinz
- 1] Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University Freiburg, Germany. [2] Department of Neuropathology, University Medical Center, Freiburg, Germany
| | - Philipp Henneke
- 1] Center of Chronic Immunodeficiency, Albert-Ludwigs-University Freiburg, Germany. [2] Center for Pediatrics and Adolescent Medicine, University Medical Center, Freiburg, Germany
| | - Hans Häcker
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gerhard C Hildebrandt
- 1] Division of Hematology and Oncology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA. [2] Division of Bone Marrow Transplantation, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah, USA. [3]
| | - Georg Häcker
- 1] Department of Medical Microbiology and Hygiene, University Medical Center, Freiburg, Germany. [2] Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University Freiburg, Germany. [3]
| | - Robert Zeiser
- 1] Department of Hematology and Oncology, University Medical Center, Freiburg, Germany. [2] Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University Freiburg, Germany. [3] Centre for Biological Signaling Studies BIOSS, Albert-Ludwigs-University Freiburg, Germany. [4]
| |
Collapse
|
42
|
Lee CS, Ryan EJ, Doherty GA. Gastro-intestinal toxicity of chemotherapeutics in colorectal cancer: The role of inflammation. World J Gastroenterol 2014; 20:3751-3761. [PMID: 24744571 PMCID: PMC3983434 DOI: 10.3748/wjg.v20.i14.3751] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 02/27/2014] [Indexed: 02/06/2023] Open
Abstract
Chemotherapy-induced diarrhea (CID) is a common and often severe side effect experienced by colorectal cancer (CRC) patients during their treatment. As chemotherapy regimens evolve to include more efficacious agents, CID is increasingly becoming a major cause of dose limiting toxicity and merits further investigation. Inflammation is a key factor behind gastrointestinal (GI) toxicity of chemotherapy. Different chemotherapeutic agents activate a diverse range of pro-inflammatory pathways culminating in distinct histopathological changes in the small intestine and colonic mucosa. Here we review the current understanding of the mechanisms behind GI toxicity and the mucositis associated with systemic treatment of CRC. Insights into the inflammatory response activated during this process gained from various models of GI toxicity are discussed. The inflammatory processes contributing to the GI toxicity of chemotherapeutic agents are increasingly being recognised as having an important role in the development of anti-tumor immunity, thus conferring added benefit against tumor recurrence and improving patient survival. We review the basic mechanisms involved in the promotion of immunogenic cell death and its relevance in the treatment of colorectal cancer. Finally, the impact of CID on patient outcomes and therapeutic strategies to prevent or minimise the effect of GI toxicity and mucositis are discussed.
Collapse
|
43
|
Abstract
Serotonin (5-hydroxytryptamine; 5-HT), a well-characterized neurotransmitter in the central nervous system, plays a crucial role in regulating mood, body temperature, sleep, appetite, and metabolism. Serotonin is synthesized in the serotonergic neuron of the central nervous system; however, approximately 90% of serotonin is synthesized and localized in the gastrointestinal (GI) tract, especially in the enterochromaffin (EC) cells. In the GI tract, serotonin mediates control over a variety of physiological functions such as contraction/relaxation of smooth muscle, and peristaltic and secretory reflexes, directly or indirectly through intrinsic primary afferent neurons. The receptors mediating the action of serotonin are mainly classified into 7 major groups known as the 5-HT1 to 5-HT7 receptors. The 5-HT3 receptor is distinguished from among the other 5-HT receptor subtypes because it is only a ligand-gated ion channel, whereas the other subtypes serve as G protein-coupled receptors. The 5-HT3 receptor, which is generally considered to be localized in the central and peripheral nervous systems, is involved in processes associated with emotion, cognition, memory, pain perception, and GI functions including secretion and motility. Recently, an increasing number of findings have provided evidence of the important role of the 5-HT3 receptor in the regulation of inflammatory and immune responses. In fact, several 5-HT3 receptor antagonists have been reported to ameliorate intestinal inflammation. Therefore, this review focuses on the role of 5-HT3 receptors in the pathogenesis of intestinal inflammation.
Collapse
Affiliation(s)
- Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University
| |
Collapse
|
44
|
Treatment withSaccharomyces boulardiireduces the inflammation and dysfunction of the gastrointestinal tract in 5-fluorouracil-induced intestinal mucositis in mice. Br J Nutr 2014; 111:1611-21. [DOI: 10.1017/s0007114513004248] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal mucositis is an important toxic side effect of 5-fluorouracil (5-FU) treatment.Saccharomyces boulardiiis known to protect from intestinal injury via an effect on the gastrointestinal microbiota. The objective of the present study was to evaluate the effect ofS. boulardiion intestinal mucositis induced by 5-FU in a murine model. Mice were divided into saline, saline (control)+5-FU or 5-FU+S. boulardii(16 × 109colony-forming units/kg) treatment groups, and the jejunum and ileum were removed after killing of mice for the evaluation of histopathology, myeloperoxidase (MPO) activity, and non-protein sulfhydryl group (mainly reduced glutathione; GSH), nitrite and cytokine concentrations. To determine gastric emptying, phenol red was administered orally, mice were killed 20 min after administration, and the absorbance of samples collected from the mice was measured by spectrophotometry. Intestinal permeability was measured by the urinary excretion rate of lactulose and mannitol following oral administration.S. boulardiisignificantly reversed the histopathological changes in intestinal mucositis induced by 5-FU and reduced the inflammatory parameters: neutrophil infiltration (control 1·73 (sem0·37) ultrastructural MPO (UMPO)/mg, 5-FU 7·37 (sem1·77) UMPO/mg and 5-FU+S. boulardii4·15 (sem0·73) UMPO/mg); nitrite concentration (control 37·00 (sem2·39) μm, 5-FU 59·04 (sem11·41) μmand 5-FU+S. boulardii37·90 (sem5·78) μm); GSH concentration (control 477·60 (sem25·25) μg/mg, 5-FU 270·90 (sem38·50) μg/mg and 5-FU+S. boulardii514·00 (sem38·64) μg/mg). Treatment with S.Boulardiisignificantly reduced the concentrations of TNF-α and IL-1β by 48·92 and 32·21 % in the jejunum and 38·92 and 61·79 % in the ileum. In addition,S. boulardiidecreased the concentrations of chemokine (C–X–C motif) ligand 1 by 5-fold in the jejunum and 3-fold in the ileum. Interestingly,S. boulardiireduced the delay in gastric emptying (control 25·21 (sem2·55) %, 5-FU 54·91 (sem3·43) % and 5-FU+S. boulardii31·38 (sem2·80) %) and induced the recovery of intestinal permeability (lactulose:mannitol ratio: control 0·52 (sem0·03), 5-FU 1·38 (sem0·24) and 5-FU+S. boulardii0·62 (sem0·03)). In conclusion,S. boulardiireduces the inflammation and dysfunction of the gastrointestinal tract in intestinal mucositis induced by 5-FU.
Collapse
|
45
|
Katsuyama M. [Superoxide-generating enzymes NADPH oxidases, potential targets of drug therapy: various mechanisms for regulation of their expression]. Nihon Yakurigaku Zasshi 2013; 142:285-90. [PMID: 24334927 DOI: 10.1254/fpj.142.285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
46
|
Kolli VK, Natarajan K, Isaac B, Selvakumar D, Abraham P. Mitochondrial dysfunction and respiratory chain defects in a rodent model of methotrexate-induced enteritis. Hum Exp Toxicol 2013; 33:1051-65. [PMID: 24347301 DOI: 10.1177/0960327113515503] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The efficacy of methotrexate (MTX), a widely used chemotherapeutic drug, is limited by its gastrointestinal toxicity and the mechanism of which is not clear. The present study investigates the possible role of mitochondrial damage in MTX-induced enteritis. Small intestinal injury was induced in Wistar rats by the administration of 7 mg kg(-1) body wt. MTX intraperitoneally for 3 consecutive days. MTX administration resulted in severe small intestinal injury and extensive damage to enterocyte mitochondria. Respiratory control ratio, the single most useful and reliable test of mitochondrial function, and 3-(4,5-dimethylthiazol-2-yll)-2,5-diphenyltetrazolium bromide reduction, a measure of cell viability were significantly reduced in all the fractions of MTX-treated rat enterocytes. A massive decrease (nearly 70%) in the activities of complexes II and IV was also observed. The results of the present study suggest that MTX-induced damage to enterocyte mitochondria may play a critical role in enteritis. MTX-induced alteration in mitochondrial structure may cause its dysfunction and decreases the activities of the electron chain complexes. MTX-induced mitochondrial damage can result in reduced adenosine triphosphate synthesis, thereby interfering with nutrient absorption and enterocyte renewal. This derangement may contribute to malabsorption of nutrients, diarrhea, and weight loss seen in patients on MTX chemotherapy.
Collapse
Affiliation(s)
- V K Kolli
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - K Natarajan
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - B Isaac
- Department of Anatomy, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - D Selvakumar
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - P Abraham
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| |
Collapse
|
47
|
Ito Y, Kinoshita M, Yamamoto T, Sato T, Obara T, Saitoh D, Seki S, Takahashi Y. A combination of pre- and post-exposure ascorbic acid rescues mice from radiation-induced lethal gastrointestinal damage. Int J Mol Sci 2013; 14:19618-35. [PMID: 24084715 PMCID: PMC3821576 DOI: 10.3390/ijms141019618] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 09/09/2013] [Accepted: 09/13/2013] [Indexed: 12/13/2022] Open
Abstract
The development of an effective therapy for radiation-induced gastrointestinal damage is important, because it is currently a major complication of treatment and there are few effective therapies available. Although we have recently demonstrated that pretreatment with ascorbic acid attenuates lethal gastrointestinal damage in irradiated mice, more than half of mice eventually died, thus indicating that better approach was needed. We then investigated a more effective therapy for radiation-induced gastrointestinal damage. Mice receiving abdominal radiation at 13 Gy were orally administered ascorbic acid (250 mg/kg/day) for three days before radiation (pretreatment), one shot of engulfment (250 mg/kg) at 8 h before radiation, or were administered the agent for seven days after radiation (post-treatment). None of the control mice survived the abdominal radiation at 13 Gy due to severe gastrointestinal damage (without bone marrow damage). Neither pretreatment with ascorbic acid (20% survival), engulfment (20%), nor post-treatment (0%) was effective in irradiated mice. However, combination therapy using ascorbic acid, including pretreatment, engulfment and post-treatment, rescued all of the mice from lethal abdominal radiation, and was accompanied by remarkable improvements in the gastrointestinal damage (100% survival). Omitting post-treatment from the combination therapy with ascorbic acid markedly reduced the mouse survival (20% survival), suggesting the importance of post-treatment with ascorbic acid. Combination therapy with ascorbic acid may be a potent therapeutic tool for radiation-induced gastrointestinal damage.
Collapse
Affiliation(s)
- Yasutoshi Ito
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8566, Japan; E-Mails: (Y.I.); (T.Y.); (T.S.); (T.O.); (Y.T.)
| | - Manabu Kinoshita
- Department of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8613, Japan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-4-2995-1541; Fax: +81-4-2996-5194
| | - Tetsuo Yamamoto
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8566, Japan; E-Mails: (Y.I.); (T.Y.); (T.S.); (T.O.); (Y.T.)
| | - Tomohito Sato
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8566, Japan; E-Mails: (Y.I.); (T.Y.); (T.S.); (T.O.); (Y.T.)
| | - Takeyuki Obara
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8566, Japan; E-Mails: (Y.I.); (T.Y.); (T.S.); (T.O.); (Y.T.)
| | - Daizoh Saitoh
- Division of Traumatology, Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8613, Japan; E-Mail:
| | - Shuhji Seki
- Department of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8613, Japan; E-Mail:
| | - Yukihiro Takahashi
- Military Medicine Research Unit, Test and Evaluation Command, Ground Self-Defense Force, 1-2-24 Ikejiri, Setagaya, Tokyo 154-8566, Japan; E-Mails: (Y.I.); (T.Y.); (T.S.); (T.O.); (Y.T.)
| |
Collapse
|
48
|
Yasuda M, Kato S, Yamanaka N, Iimori M, Matsumoto K, Utsumi D, Kitahara Y, Amagase K, Horie S, Takeuchi K. 5-HT₃ receptor antagonists ameliorate 5-fluorouracil-induced intestinal mucositis by suppression of apoptosis in murine intestinal crypt cells. Br J Pharmacol 2013; 168:1388-400. [PMID: 23072534 DOI: 10.1111/bph.12019] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 09/23/2012] [Accepted: 10/02/2012] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Chemotherapeutic agents, including 5-fluorouracil (5-FU), frequently cause intestinal mucositis resulting in severe diarrhoea and morphological mucosal damage. 5-HT₃ receptor antagonists are clinically effective in the treatment of nausea and emesis during cancer chemotherapy. Therefore we here have examined the effects of 5-HT₃ receptor antagonists on 5-FU-induced intestinal mucositis in mice. EXPERIMENTAL APPROACH Intestinal mucositis was induced in male C57BL/6 mice by daily administration of 5-FU (50 mg·kg⁻¹) for 5 days. Effects of 5-HT₃ receptor antagonists, ramosetron (0.01-0.1 mg·kg⁻¹) and ondansetron (5 mg·kg⁻¹), on the accompanying histology, cytokine production and apoptosis were assessed. KEY RESULTS Continuous administration of 5-FU to mice caused severe intestinal mucositis, which was histologically characterized by the shortening of villi and destruction of intestinal crypts, accompanied by body weight loss and diarrhoea. Daily ramosetron administration dose-dependently reduced the severity of intestinal mucositis, body weight loss and diarrhoea. Similar beneficial effects were observed with ondansetron. The number of apoptotic, caspase-3- and caspase-8-activated cells increased 24 h after the first 5-FU administration, and these responses were reduced by ramosetron. The up-regulation of TNF-α, IL-1β and IL-6 following 5-FU treatment was also attenuated by ramosetron. CONCLUSIONS AND IMPLICATIONS 5-HT₃ receptor antagonists ameliorated 5-FU-induced intestinal mucositis in mice, and this action could result from suppression of apoptotic responses in the intestinal crypt cells via inhibition of cytokine expression. Thus, 5-HT₃ receptor antagonists may be useful for preventing not only nausea and emesis but also intestinal mucositis during 5-FU chemotherapy.
Collapse
Affiliation(s)
- M Yasuda
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Oxidative Stress, Nox Isoforms and Complications of Diabetes—Potential Targets for Novel Therapies. J Cardiovasc Transl Res 2012; 5:509-18. [DOI: 10.1007/s12265-012-9387-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 06/06/2012] [Indexed: 01/02/2023]
|