1
|
Zhou Y, Han W, Feng Y, Wang Y, Sun T, Xu J. Microbial metabolites affect tumor progression, immunity and therapy prediction by reshaping the tumor microenvironment (Review). Int J Oncol 2024; 65:73. [PMID: 38847233 PMCID: PMC11173369 DOI: 10.3892/ijo.2024.5661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/30/2024] [Indexed: 06/12/2024] Open
Abstract
Several studies have indicated that the gut microbiome and tumor microbiota may affect tumors. Emerging metabolomics research illustrates the need to examine the variations in microbial metabolite composition between patients with cancer and healthy individuals. Microbial metabolites can impact the progression of tumors and the immune response by influencing a number of mechanisms, including modulation of the immune system, cancer or immune‑related signaling pathways, epigenetic modification of proteins and DNA damage. Microbial metabolites can also alleviate side effects and drug resistance during chemotherapy and immunotherapy, while effectively activating the immune system to exert tumor immunotherapy. Nevertheless, the impact of microbial metabolites on tumor immunity can be both beneficial and harmful, potentially influenced by the concentration of the metabolites or the specific cancer type. The present review summarizes the roles of various microbial metabolites in different solid tumors, alongside their influence on tumor immunity and treatment. Additionally, clinical trials evaluating the therapeutic effects of microbial metabolites or related microbes on patients with cancer have been listed. In summary, studying microbial metabolites, which play a crucial role in the interaction between the microbiota and tumors, could lead to the identification of new supplementary treatments for cancer. This has the potential to improve the effectiveness of cancer treatment and enhance patient prognosis.
Collapse
Affiliation(s)
- Yuhang Zhou
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
| | - Wenjie Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
| | - Yun Feng
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
| | - Yue Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
| | - Tao Sun
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Oncology Medicine, Key Laboratory of Liaoning Breast Cancer Research, Shenyang, Liaoning 110042, P.R. China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
| | - Junnan Xu
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
2
|
Liang H, Fan X, Cheng H, Ma X, Sun Y, Nan F, Zhou J, Shu P, Zhang W, Zuo F, Nakatsukasa H, Zhang D. CPT-11 mitigates autoimmune diseases by suppressing effector T cells without affecting long-term anti-tumor immunity. Cell Death Discov 2024; 10:218. [PMID: 38704362 PMCID: PMC11069576 DOI: 10.1038/s41420-024-01983-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
The incidence of autoimmune diseases has significantly increased over the past 20 years. Excessive host immunoreactions and disordered immunoregulation are at the core of the pathogenesis of autoimmune diseases. The traditional anti-tumor chemotherapy drug CPT-11 is associated with leukopenia. Considering that CPT-11 induces leukopenia, we believe that it is a promising drug for the control of autoimmune diseases. Here, we show that CPT-11 suppresses T cell proliferation and pro-inflammatory cytokine production in healthy C57BL/6 mice and in complete Freund's adjuvant-challenged mice. We found that CPT-11 effectively inhibited T cell proliferation and Th1 and Th17 cell differentiation by inhibiting glycolysis in T cells. We also assessed CPT-11 efficacy in treating autoimmune diseases in models of experimental autoimmune encephalomyelitis and psoriasis. Finally, we proved that treatment of autoimmune diseases with CPT-11 did not suppress long-term immune surveillance for cancer. Taken together, these results show that CPT-11 is a promising immunosuppressive drug for autoimmune disease treatment.
Collapse
Affiliation(s)
- Hantian Liang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinzou Fan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hao Cheng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiao Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yutong Sun
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fang Nan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jingyang Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Panyin Shu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wei Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fengqiong Zuo
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Hiroko Nakatsukasa
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8675, Japan
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
3
|
Li Z, Ke X, Zuo D, Wang Z, Fang F, Li B. New Insights into the Relationship between Gut Microbiota and Radiotherapy for Cancer. Nutrients 2022; 15:nu15010048. [PMID: 36615706 PMCID: PMC9824372 DOI: 10.3390/nu15010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
Cancer is the second most common cause of death among humans in the world, and the threat that it presents to human health is becoming more and more serious. The mechanisms of cancer development have not yet been fully elucidated, and new therapies are changing with each passing day. Evidence from the literature has validated the finding that the composition and modification of gut microbiota play an important role in the development of many different types of cancer. The results also demonstrate that there is a bidirectional interaction between the gut microbiota and radiotherapy treatments for cancer. In a nutshell, the modifications of the gut microbiota caused by radiotherapy have an effect on tumor radiosensitivity and, as a result, affect the efficacy of radiotherapy and show a certain radiation toxicity, which leads to numerous side effects. What is of new research significance is that the "gut-organ axis" formed by the gut microbiota may be one of the most interesting potential mechanisms, although the relevant research is still very limited. In this review, we combine new insights into the relationship between the gut microbiota, cancer, and radiotherapy. Based on our current comprehensive understanding of this relationship, we give an overview of the new cancer treatments based on the gut microbiota.
Collapse
Affiliation(s)
- Zhipeng Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Xiyang Ke
- Key Laboratory of Carcinogenesis and Translational Research, Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Ministry of Education, Beijing 100142, China
| | - Dan Zuo
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Fang Fang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: ; Tel.: +86-431-85619455
| | - Bo Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- School of Public Health, Jilin University, Changchun 130021, China
| |
Collapse
|
4
|
Liao C, Wang D, Qin S, Zhang Y, Chen J, Xu R, Xu F, Zhang P. Inflammatory-Dependent Bidirectional Effect of Bile Acids on NLRP3 Inflammasome and Its Role in Ameliorating CPT-11-Induced Colitis. Front Pharmacol 2022; 13:677738. [PMID: 35712724 PMCID: PMC9193974 DOI: 10.3389/fphar.2022.677738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/12/2022] [Indexed: 12/08/2022] Open
Abstract
Irinotecan (CPT-11) in combination with 5-fluorouracil and leucovorin is a first-line chemotherapy regimen for the treatment of colorectal cancer; however, its clinical application is limited by the dose-limiting gastrointestinal toxicity of colitis. In our previous studies, several bile acids (BAs) were found significantly elevated in the colon of the CPT-11-induced rat colitis model. On the other hand, NLRP3 inflammasome has been reported to play important roles in mediating colitis. Interestingly, BA was stated to activate the NLRP3 inflammasome in some studies, while in some other reports, it showed an inhibitory effect. We assumed that the inflammatory status in different circumstances might have contributed to the controversial findings. In this study, we first discovered, under non-inflammatory conditions, that supplementing BA could activate the NLRP3 inflammasome in THP-1-differentiated macrophages and promote inflammation. In lipopolysaccharide (LPS)-induced inflammatory macrophages, however, BA inhibited the NLRP3 inflammasome and reduced inflammation. Further experiments demonstrated that Takeda G protein-coupled receptor 5 (TGR5) is essential in mediating the inhibitory effect of BA, while phospho-SP1 (p-SP1) is key to the activation. Furthermore, we applied the above findings to ameliorate CPT-11-caused colitis in rats by inhibiting SP1 with mithramycin A (MitA) or activating TGR5 using oleanolic acid (OA). Our findings may shed light on the discovery of effective interventions for reducing dose-limiting chemotherapy-induced colitis.
Collapse
Affiliation(s)
- Chuyao Liao
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Di Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Siyuan Qin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Ying Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Jie Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Ruijie Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Zhou Q, Wang Y, Gu Y, Li J, Wang H, Leng J, Li W, Yu Z, Hu G, Ma RCW, Fang ZZ, Yang X, Jiang G. Genetic variants associated with beta-cell function and insulin sensitivity potentially influence bile acid metabolites and gestational diabetes mellitus in a Chinese population. BMJ Open Diabetes Res Care 2021; 9:9/1/e002287. [PMID: 34518156 PMCID: PMC8438732 DOI: 10.1136/bmjdrc-2021-002287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/17/2021] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION To investigate associations between genetic variants related to beta-cell (BC) dysfunction or insulin resistance (IR) in type 2 diabetes (T2D) and bile acids (BAs), as well as the risk of gestational diabetes mellitus (GDM). RESEARCH DESIGN AND METHODS We organized a case-control study of 230 women with GDM and 217 without GDM nested in a large prospective cohort of 22 302 Chinese women in Tianjin, China. Two weighted genetic risk scores (GRSs), namely BC-GRS and IR-GRS, were established by combining 39 and 23 single nucleotide polymorphisms known to be associated with BC dysfunction and IR, respectively. Regression and mediation analyses were performed to evaluate the relationship of GRSs with BAs and GDM. RESULTS We found that the BC-GRS was inversely associated with taurodeoxycholic acid (TDCA) after adjustment for confounders (Beta (SE)=-0.177 (0.048); p=2.66×10-4). The BC-GRS was also associated with the risk of GDM (OR (95% CI): 1.40 (1.10 to 1.77); p=0.005), but not mediated by TDCA. Compared with individuals in the low tertile of BC-GRS, the OR for GDM was 2.25 (95% CI 1.26 to 4.01) in the high tertile. An interaction effect of IR-GRS with taurochenodeoxycholic acid (TCDCA) on the risk of GDM was evidenced (p=0.005). Women with high IR-GRS and low concentration of TCDCA had a markedly higher OR of 14.39 (95% CI 1.59 to 130.16; p=0.018), compared with those with low IR-GRS and high TCDCA. CONCLUSIONS Genetic variants related to BC dysfunction and IR in T2D potentially influence BAs at early pregnancy and the development of GDM. The identification of both modifiable and non-modifiable risk factors may facilitate the identification of high-risk individuals to prevent GDM.
Collapse
Affiliation(s)
- Qiulun Zhou
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ying Wang
- The Second School of Clinical Medicine, Key Laboratory of 3D Printing Technology in Stomatology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yuqin Gu
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Junhong Leng
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhong-Ze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Dai LS, Tian HF, Hang Y, Wen CW, Huang YH, Wang BF, Hu JW, Xu JP, Deng MJ. 1 H NMR-based metabonomic evaluation of the pesticides camptothecin and matrine against larvae of Spodoptera litura. PEST MANAGEMENT SCIENCE 2021; 77:208-216. [PMID: 32677739 DOI: 10.1002/ps.6009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/10/2020] [Accepted: 07/17/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Camptothecin (CPT) and matrine (MAT) have potential as botanical pesticides against several pest species. However, the mechanisms of metabolic and physiological changes in pests induced by CPT and MAT are unknown. In this study, a toxicological test, an NMR-based metabolomic study, an enzymatic test, and an RT quantitative PCR (RT-qPCR) experiment were all conducted to examine the effect of CPT and MAT on Spodoptera litura. RESULTS CPT (0.5-1%) exerted high toxicity against larvae of S. litura and caused growth stagnation and high mortality of larvae. A variety of metabolites were significantly influenced by 0.5% CPT, including several energy-related metabolites such as trehalose, lactate, succinate, citrate, malate, and fumarate. In contrast, MAT showed low toxicity against larvae and induced almost no changes in hemolymph metabolites of S. litura. Enzymatic tests showed that trehalase activity was significantly decreased in larvae after feeding with 0.5% CPT. RT-qPCR showed that the transcription levels of alanine aminotransferase, malate dehydrogenase, and isocitrate dehydrogenase were decreased while lactate dehydrogenase was increased in the 0.5% CPT-treated group. CONCLUSIONS These data indicate that one of the important mechanisms of CPT against S. litura larvae is via the inhibition of trehalose hydrolysis and glycolysis. Our findings also suggest that CPT exhibits a stronger toxicological effect than MAT against S. litura, which provides basic information for the application of CPT in the control of S. litura or other lepidoptera pests.
Collapse
Affiliation(s)
- Li-Shang Dai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui-Fei Tian
- School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yang Hang
- Biotechnology Center of Anhui Agricultural University, Anhui Agricultural University, Hefei, China
| | - Chao-Wei Wen
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ying-Hao Huang
- Renji College, Wenzhou Medical University, Wenzhou, China
| | - Bin-Feng Wang
- School of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jing-Wei Hu
- Biotechnology Center of Anhui Agricultural University, Anhui Agricultural University, Hefei, China
| | - Jia-Ping Xu
- School of Life Sciences, Anhui Agricultural University, Hefei, China
| | - Ming-Jie Deng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Irinotecan-induced intestinal mucositis in mice: a histopathological study. Cancer Chemother Pharmacol 2020; 87:327-336. [PMID: 33130913 DOI: 10.1007/s00280-020-04186-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Intestinal mucositis is an important adverse effect of antineoplastic therapy, which remains without adequate treatment. The present study aimed to carry out a complete evaluation of the histopathological changes during irinotecan-induced intestinal mucositis, using the protocol most found in the pharmacological reports nowadays to better understand irinotecan toxicity and support future studies on drug discovery. METHODS Intestinal mucositis was induced by treating swiss mice for 4 days with irinotecan (75 mg/kg, i.p.). After 72 h post irinotecan, the mice were sacrificed and the small intestine and colon were excised to performed histological analysis by stained tissue with hematoxylin/eosin (H&E). RESULTS Histoarchitecture loss, villus/crypt ratio reduction, atrophy of the muscular layer, hypertrophy in the submucosal and mucous layers, ruptures in the epithelium, as well as extent cellular infiltrate and presence of micro abscesses and the fusion of the crypts were observed in the histological analysis. Moreover, duodenum and colon had increased intraepithelial lymphocytes and mitotic figures. However, submucosal ganglia were decreased in the duodenum and increased in the colon. CONCLUSIONS The data obtained in the present study provides new evidence that irinotecan-induced intestinal mucositis highly affects small intestine and colon, further contributing to establish criteria in light of the histopathological changes induced by irinotecan during intestinal mucositis and facilitating inter-study comparisons.
Collapse
|
8
|
Wang T, Lin S, Liu R, Li H, Liu Z, Xu H, Li Q, Bi K. Acute lung injury therapeutic mechanism exploration for Chinese classic prescription Qingzao Jiufei Decoction by UFLC-MS/MS quantification of bile acids, fatty acids and eicosanoids in rats. J Pharm Biomed Anal 2020; 189:113463. [PMID: 32688210 DOI: 10.1016/j.jpba.2020.113463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/02/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is a common and complex inflammatory disease, which has been reasonably associated with carboxyl-containing metabolites in our preliminary non-targeted metabolomic strategy. Qingzao Jiufei Decoction (QZJFD), a classic prescription, is widely used in the treatment of pulmonary inflammatory injuries. Successively, in this targeted project, to fill in the research gap and exposit the therapeutic mechanism of QZJFD on ALI, considering the structure similarity and bioactivity correlation, 21 bile acids, 11 fatty acids and 19 eicosanoids were profiled simultaneously in plasma, lung, bronchoalveolar lavage fluid, spleen and feces from rats utilizing a novel ultraperformance liquid chromatography-mass spectrometry approach. As a result, potential biomarkers and ALI characteristic metabolomic spectrums were obtained to distinguish different physical states using discriminative similarity threshold as 0.65 for clinical application. After treatment with QZJFD, obvious reversing ability for various biomarker levels was observed in different bio-samples, providing insights into the systemic intervention of QZJFD on ALI by regulating bile acid synthesis, fatty acid synthesis and eicosanoid metabolism. Conclusively, this investigation represented more information on the comprehensive therapeutic action of QZJFD on ALI involving with multi-targets and multi-pathways for clinical application and traditional Chinese medicine modernization.
Collapse
Affiliation(s)
- Tianyang Wang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China
| | - Song Lin
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, Heilongjiang Province, 161006, China
| | - Ran Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China
| | - Hua Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China
| | - Zihan Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China
| | - Huarong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China
| | - Qing Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
9
|
da Silva Ferreira AR, Wardill HR, Tissing WJ, Harmsen HJ. Pitfalls and novel experimental approaches to optimize microbial interventions for chemotherapy-induced gastrointestinal mucositis. Curr Opin Support Palliat Care 2020; 14:127-134. [PMID: 32324645 PMCID: PMC7259380 DOI: 10.1097/spc.0000000000000497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW There is a growing number of studies implicating gut dysbiosis in mucositis development. However, few studies have shed light on the causal relationship limiting translational potential. Here, we detail the key supportive evidence for microbial involvement, candidate mechanisms by which the microbiome may contribute to mucositis and emerging approaches to model host-microbe interactions with clinical relevance and translational potential. RECENT FINDINGS Synthesis of existing clinical data demonstrate that modulating the microbiome drastically alters the development and severity of mucositis, providing a strong rationale for its involvement. Review of the literature revealed potential microbiome-dependent mechanisms of mucosal injury including altered drug metabolism, bile acid synthesis and regulation of the intestinal barrier. Current studies are limited in their mechanistic insight due to cross-sectional and would benefit from longitudinal analyses and baseline phenotyping. SUMMARY The causative role of the microbiome in mucositis development remains unclear. Future studies must adopt comprehensive microbial analyses with functional assessment, and utilize emerging ex-vivo models to interrogate host-microbe interactions in mucositis.
Collapse
Affiliation(s)
| | - Hannah R. Wardill
- Department of Pediatrics Oncology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Wim J.E. Tissing
- Department of Pediatrics Oncology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | |
Collapse
|
10
|
Jiawei Xianglian Decoction (JWXLD), a Traditional Chinese Medicine (TCM), Alleviates CPT-11-Induced Diarrhea in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7901231. [PMID: 32256654 PMCID: PMC7091529 DOI: 10.1155/2020/7901231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 01/19/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
Abstract
Irinotecan (CPT-11) is used for therapy of various cancers. However, it has several serious adverse reactions such as diarrhea which is caused by SN-38, the active form of CPT-11. This study aimed to evaluate the effectiveness of Jiawei xianglian decoction (JWXLD), which has been widely used for the treatment of diarrhea in China. In this study, a mouse model with delayed diarrhea was generated by CPT-11. Real-time PCR and enzyme-linked immunosorbent assay (ELISA) were performed to explore intestinal microflora and inflammatory cytokine. Hematoxylin and eosin (H&E) staining was used to analyze tissue morphology. We found that 0.12, 0.23, and 0.46 g JWXLD significantly reduced the severity of CPT-11-induced diarrhea. The levels of Lactobacillus (Lacto) and Bifidobacterium (Bifid) were significantly downregulated by CPT-11, and these effects can be reversed by JWXLD treatment. Furthermore, JWXLD was observed to decrease the activity of β-glucuronidase (β-GD). Histopathological data showed that CPT-11 induced severe intestinal mucosal injury, which was characterized as grade 6, and JWXLD significantly alleviated the injury. In addition, CPT-11 increased the productions of tumor necrosis factor-alpha (TNF-α), tumor necrosis factor-beta (TNF-β), interleukin-6 (IL-6), and interleukin-1 (IL-1), but decreased interleukin-15 (IL-15), interleukin-7 (IL-7), and uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1). In conclusion, JWXLD can counteract these effects caused by CPT-11 treatment. JWXLD could alleviate CPT-11-induced diarrhea.
Collapse
|
11
|
Wang T, Lin S, Liu R, Li H, Liu Z, Zhang X, Xu H, Li Q, Bi K. Metabolomic profile perturbations of serum, lung, bronchoalveolar lavage fluid, spleen and feces in LPS-induced acute lung injury rats based on HPLC-ESI-QTOF-MS. Anal Bioanal Chem 2020; 412:1215-1234. [PMID: 31940090 DOI: 10.1007/s00216-019-02357-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/03/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
Acute lung injury (ALI) is a clinically common and serious disease, underscoring the urgent need for clarification of its pathogenesis. According to traditional Chinese medicine (TCM) theories on the "lung-spleen-intestine axis" and its correlation with ALI, a high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (HPLC-QTOF-MS) metabolomic platform was applied to identify biomarkers from five bio-samples of control and model rats challenged with intratracheally administered lipopolysaccharide (LPS) based on multivariate mathematical statistical analysis. As a result, 19, 24, 24, 15 and 29 altered metabolites were identified in serum, lung, bronchoalveolar lavage fluid (BALF), spleen and feces samples, respectively. Metabolic pathway analysis showed that linoleic acid, sphingolipid, glycerophospholipid and bile acid metabolism pathways were mainly altered by ALI. Additionally, ROC curves were applied to assess the specificity and sensitivity of the biomarkers. ALI characteristic metabolomic spectra were then established to differentiate the control from the model group with a similarity discriminative threshold of 0.7. Additionally, to compare the metabolomic profiles of the five bio-samples and establish metabolic similarities and differences among them, correlation analysis was conducted in order to delineate an objective law of endogenous linkage along the lung-spleen-intestine axis. Therefore, this study provides insights into the mechanisms involved in ALI from a metabolomics perspective, which can be applied in characterization of the mechanism and early disease detection. Graphical abstract.
Collapse
Affiliation(s)
- Tianyang Wang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Song Lin
- Basic Medical Science College, Qiqihar Medical University, Qiqihar, 161006, Heilongjiang, China
| | - Ran Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Hua Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Zihan Liu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Xinnong Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Huarong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Qing Li
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China
| | - Kaishun Bi
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
12
|
Wu CH, Ko JL, Liao JM, Huang SS, Lin MY, Lee LH, Chang LY, Ou CC. D-methionine alleviates cisplatin-induced mucositis by restoring the gut microbiota structure and improving intestinal inflammation. Ther Adv Med Oncol 2019; 11:1758835918821021. [PMID: 30792823 PMCID: PMC6376546 DOI: 10.1177/1758835918821021] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023] Open
Abstract
Background There are close links between chemotherapy-induced intestinal mucositis and microbiota dysbiosis. Previous studies indicated that D-methionine was an excellent candidate for a chemopreventive agent. Here, we investigated the effects of D-methionine on cisplatin-induced mucositis. Materials and methods Male Wistar rats (176-200 g, 6 weeks old) were given cisplatin (5 mg/kg) and treated with D-methionine (300 mg/kg). Histopathological, digestive enzymes activity, oxidative/antioxidant status, proinflammatory/anti-inflammatory cytokines in intestinal tissues were measured. Next-generation sequencing technologies were also performed to investigate the gut microbial ecology. Results D-methionine administration increased villus length and crypt depth and improved digestive enzyme (leucine aminopeptidase, sucrose and alkaline phosphatase) activities in the brush-border membrane of cisplatin-treated rats (p < 0.05). Furthermore, D-methionine significantly attenuated oxidative stress and inflammatory reaction and increased interleukin-10 levels in cisplatin-induced intestinal mucositis (p < 0.05). Cisplatin administration resulted in high relative abundances of Deferribacteres and Proteobacteria and a low diversity of the microbiota when compared with control groups, D-methionine only and cisplatin plus D-methionine. Cisplatin markedly increased comparative abundances of Bacteroides caccae, Escherichia coli, Mucispirillum schaedleri, Bacteroides uniformis and Desulfovibrio C21-c20, while Lactobacillus was almost completely depleted, compared with the control group. There were higher abundances of Lactobacillus, Lachnospiraceae, and Clostridium butyrium in cisplatin plus D-methionine rats than in cisplatin rats. D-methionine treatment alone significantly increased the number of Lactobacillus reuteri. Conclusion D-methionine protects against cisplatin-induced intestinal damage through antioxidative and anti-inflammatory effects. By enhancing growth of beneficial bacteria (Lachnospiraceae and Lactobacillus), D-methionine attenuates gut microbiome imbalance caused by cisplatin and maintains gut homeostasis.
Collapse
Affiliation(s)
- Cheng-Hsi Wu
- Department of Family Medicine, Jen-Ai Hospital, Dali, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiuan-Miaw Liao
- Department of Physiology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shiang-Suo Huang
- Department of Pharmacology and Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Meei-Yn Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ling-Hui Lee
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Li-Yu Chang
- Department of Nursing, Jen-Ai Hospital, Dali, Taichung, Taiwan
| | - Chu-Chyn Ou
- School of Nutrition, Chung Shan Medical University, 110, Sec. 1, Chien-Kuo N. Road, Taichung 40203, Taiwan
| |
Collapse
|
13
|
Takahashi S, Tanaka N, Fukami T, Xie C, Yagai T, Kim D, Velenosi TJ, Yan T, Krausz KW, Levi M, Gonzalez FJ. Role of Farnesoid X Receptor and Bile Acids in Hepatic Tumor Development. Hepatol Commun 2018; 2:1567-1582. [PMID: 30556042 PMCID: PMC6287584 DOI: 10.1002/hep4.1263] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/09/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer deaths worldwide, and an association between altered bile acid (BA) metabolism, down‐regulation of farnesoid X receptor (FXR), which is a master regulator of BA metabolism, and hepatocarcinogenesis has been documented. While global FXR deficiency in mice results in spontaneous HCC with aging, the contribution of tissue‐specific FXR deficiency to hepatocarcinogenesis remains unclear. In this study, the prevalence of hepatic tumors, expression of genes related to tumorigenesis, and serum/liver BA levels were compared among male whole‐body Fxr‐null, hepatocyte‐specific Fxr‐null (Fxr∆Hep), and enterocyte‐specific Fxr‐null (Fxr∆IE) mice at the age of 3, 14, and 20 months. More than 90% of 20‐month‐old whole‐body Fxr‐null mice had hepatic tumors with enhanced hepatic expression of myelocytomatosis oncogene (Myc) and cyclin‐dependent kinase 4 (Cdk4) messenger RNAs (mRNAs) and elevated serum taurocholate (TCA) and tauromuricholate (TMCA) and their respective unconjugated derivatives. The incidence of hepatic tumors was significantly lower in Fxr∆Hep and Fxr∆IE mice (20% and 5%, respectively), and the increases in Myc and Cdk4 mRNA or serum BA concentrations were not detected in these mice compared to Fxrfloxed [fl]/fl mice; a similar tendency was observed in 14‐month‐old mice. However, increased hepatic c‐Myc protein expression was found only in Fxr‐null mice at the age of 3, 14, and 20 months. Treatment with TCA induced Myc expression in Fxr‐null cultured primary mouse hepatocytes but not in wild‐type (WT) mouse hepatocytes, demonstrating that the combination of hepatocyte FXR disruption with elevated TCA is required for Myc induction and ensuing age‐dependent hepatocarcinogenesis in Fxr‐null mice. Conclusion: There is a relatively low risk of hepatic tumors by inhibition of FXR in enterocytes, likely due to the lack of increased TCA and Myc induction.
Collapse
Affiliation(s)
- Shogo Takahashi
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD.,Department of Biochemistry and Molecular and Cellular Biology Georgetown University Washington DC
| | - Naoki Tanaka
- Department of Metabolic Regulation Shinshu University School of Medicine Matsumoto Japan.,International Research Center for Agricultural Food Industry Shinshu University Matsumoto Japan
| | - Tatsuki Fukami
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD.,Present address: Department of Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences Kanazawa University Kanazawa Japan
| | - Cen Xie
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| | - Tomoki Yagai
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| | - Donghwan Kim
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| | - Thomas J Velenosi
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| | - Tingting Yan
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| | - Kristopher W Krausz
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology Georgetown University Washington DC
| | - Frank J Gonzalez
- Laboratory of Metabolism National Cancer Institute, National Institutes of Health Bethesda MD
| |
Collapse
|
14
|
Li J, Huo X, Cao YF, Li SN, Du Z, Shao P, Leng J, Zhang C, Sun XY, Ma RCW, Fang ZZ, Yang X. Bile acid metabolites in early pregnancy and risk of gestational diabetes in Chinese women: A nested case-control study. EBioMedicine 2018; 35:317-324. [PMID: 30120081 PMCID: PMC6161472 DOI: 10.1016/j.ebiom.2018.08.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Background Bile acid metabolism plays an important role in metabolism but it is uncertain whether bile acid metabolites in early pregnancy are associated with risk of gestational diabetes mellitus (GDM). Methods We organized a 1:1 case-control study nested in a prospective cohort of 22,302 pregnant women recruited from 2010 to 2012 in China: 243 women with GDM were matched with 243 non-GDM controls on age (±1 year). Conditional logistic regression and restricted cubic spline were used to examine full-range associations of bile acid metabolites with GDM. Findings All the 9 detectable bile acids were inversely associated with the risk of GDM, among them, 8 in nonlinear and one in largely linear manners in multivariable analysis. Glycoursodeoxycholic acid (GUDCA) at ≤0.07 nmol/mL and deoxycholic acid (DCA) at ≤0.28 nmol/mL had threshold effects and their decreasing levels below the cutoff points were associated with rapid rises in the risk of GDM. In traditional risk factor model, the stepwise procedure identified that GUDCA ≤ 0.07 nmol/mL and DCA ≤ 0.280 nmol/mL were still significant (OR: 6.84, 95%CI: 1.10–42.48 & 2.06, 1.26–3.37), while other bile acids were not. Inclusion of the two bile acids in the model increased the area under operating characteristic's curve from 0.69 to 0.76 (95% CI: 0.71–0.80) (P < .05). Interpretation Serum GUDCA ≤ 0.07 nmol/mL and DCA ≤ 0.28 nmol/mL in early pregnancy were independently associated with increased risk of GDM in Chinese pregnant women. Funding Talent Recruitment Scheme grant of Tianjin Medical University and National Key Research and Development Program, etc.
Collapse
Affiliation(s)
- Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoxu Huo
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yun-Feng Cao
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China; RSKT Biopharma Inc, Dalian, Liaoning, China
| | - Sai-Nan Li
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zuo Du
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ping Shao
- Tianjin Women and Children's Health Center, Tianjin, China
| | - Junhong Leng
- Tianjin Women and Children's Health Center, Tianjin, China
| | - Cuiping Zhang
- Tianjin Women and Children's Health Center, Tianjin, China
| | - Xiao-Yu Sun
- Key Laboratory of Liaoning Tumor Clinical Metabolomics (KLLTCM), Jinzhou, Liaoning, China; RSKT Biopharma Inc, Dalian, Liaoning, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhong-Ze Fang
- Department of Toxicology, School of Public Health, Tianjin Medical University, Tianjin, China.
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
15
|
Adoptive Induced Antigen-Specific Treg Cells Reverse Inflammation in Collagen-Induced Arthritis Mouse Model. Inflammation 2017; 41:485-495. [DOI: 10.1007/s10753-017-0704-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Zhang P, Zhang L, Zhang Y, Mao L, Jiang H. Substitutions in Spodoptera exigua topoisomerase I modulate its relaxation activity and camptothecin sensitivity. PEST MANAGEMENT SCIENCE 2017; 73:1179-1186. [PMID: 27643798 DOI: 10.1002/ps.4440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/25/2016] [Accepted: 09/12/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Topoisomerase I (Top I) is referred as the cellular target of the camptothecins (CPTs) which are now being explored as potential pesticides for insect control. Three amino acid substitutions, including L530P, A653T and S729T, in Top Is of insects were found in our previous studies. In order to investigate the effect of these three substitutions, a comparative analysis was conducted between the wild-type and mutant Top Is in Spodoptera exigua Hübner. RESULTS The optimal salt concentration of A653T and S729T was 150 mm, which is consistent with that of the wild-type Top I, while the mutant L530P showed maximum relaxation activity at a lower KCl concentration (100 mm). The mutated L530P and A653T Top Is showed a higher relaxation efficiency owing to an increased relaxation velocity toward the negatively supercoiled plasmid pBR322 DNA, which rendered L530P and A653T resistant to CPTs, whereas mutant S729T exhibited sensitivity to CPTs as a result of a decreased relaxation activity toward plasmid pBR322 DNA. CONCLUSIONS These results suggested that the polymorphism in Top I of insects was related to the biological activity of CPTs, which provided the basic information for reasonable usage of CPTs to control insect pests. © 2016 Society of Chemical Industry.
Collapse
Affiliation(s)
- Pei Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lan Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanning Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liangang Mao
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongyun Jiang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
17
|
Rattray NJW, Charkoftaki G, Rattray Z, Hansen JE, Vasiliou V, Johnson CH. Environmental influences in the etiology of colorectal cancer: the premise of metabolomics. CURRENT PHARMACOLOGY REPORTS 2017; 3:114-125. [PMID: 28642837 PMCID: PMC5475285 DOI: 10.1007/s40495-017-0088-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW In this review we discuss how environmental exposures predominate the etiology of colorectal cancer (CRC). With CRC being a personalized disease influenced by genes and environment, our goal was to explore the role metabolomics can play in identifying exposures, assessing the interplay between co-exposures, and the development of personalized therapeutic interventions. RECENT FINDINGS Approximately 10 % of CRC cases can be explained by germ-line mutations, whereas the prevailing majority are caused by an initiating exposure event occurring decades prior to diagnosis. Recent research has shown that dietary metabolites are linked to a procarcinogenic or protective environment in the colon which is modulated by the microbiome. In addition, excessive alcohol has been shown to increase the risk of CRC and is dependent on diet (folate), the response of microbiome, and genetic polymorphisms within the folate and alcohol metabolic pathways. Metabolomics can not only be used to identify this modulation of host metabolism, which could affect the progression of the tumors but also response to targeted therapeutics. SUMMARY This review highlights the current understanding of the multifaceted etiology and mechanisms of CRC development but also highlights where the field of metabolomics can contribute to a greater understanding of environmental exposure in CRC.
Collapse
Affiliation(s)
- Nicholas J. W. Rattray
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| | - Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| | - Zahra Rattray
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Yale University, CT, USA 06520
| | - James E. Hansen
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Yale University, CT, USA 06520
- Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA 06520
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| |
Collapse
|
18
|
Shi AX, Zhou Y, Zhang XY, Zhao YS, Qin HY, Wang YP, Wu XA. Irinotecan-induced bile acid malabsorption is associated with down-regulation of ileal Asbt ( Slc10a2 ) in mice. Eur J Pharm Sci 2017; 102:220-229. [DOI: 10.1016/j.ejps.2017.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 12/17/2022]
|
19
|
Wang X, Cui DN, Dai XM, Wang J, Zhang W, Zhang ZJ, Xu FG. HuangQin Decoction Attenuates CPT-11-Induced Gastrointestinal Toxicity by Regulating Bile Acids Metabolism Homeostasis. Front Pharmacol 2017; 8:156. [PMID: 28424615 PMCID: PMC5371663 DOI: 10.3389/fphar.2017.00156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 03/10/2017] [Indexed: 01/01/2023] Open
Abstract
Irinotecan (CPT-11) is a potent chemotherapeutic agent, however, its clinical usage is often limited by the induction of severe gastrointestinal (GI) toxicity, especially late-onset diarrhea. HuangQin Decoction (HQD), commonly used for the treatment of GI ailments, has been proved could significantly ameliorate the intestinal toxicity of CPT-11. To reveal the mechanisms of CPT-11-induced toxicity and the modulation effects of HQD, a previous untargeted metabolomics study was performed and the results indicated that HQD may protect the GI tract by altering the metabolism of bile acids (BAs). Nevertheless, the untargeted assays are often less sensitive and/or efficient. In order to further confirm our previous findings, here in this paper, serum and tissues metabolic profiles of 17 BAs were analyzed using liquid chromatography-tandem mass spectrometry based targeted metabolomics. The results indicated that serum and tissues levels of most BAs were significantly decreased after CPT-11 administration, except some hydrophobic BAs. Co-treatment with HQD could markedly attenuate CPT-11-induced GI toxicity and reverse the alterations of hydrophobic BAs. Despite the fact that the BAs pool size remained unchanged, the balance of BAs had shifted leading to decreased toxicity after HQD treatment. The present study demonstrated for the first time that the precise interaction between HQD, CPT-11-induced intestinal toxicity and BAs’ homeostasis.
Collapse
Affiliation(s)
- Xu Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education (MOE), China Pharmaceutical UniversityNanjing, China.,State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Dong-Ni Cui
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education (MOE), China Pharmaceutical UniversityNanjing, China.,State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Xiao-Min Dai
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education (MOE), China Pharmaceutical UniversityNanjing, China.,State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Jing Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education (MOE), China Pharmaceutical UniversityNanjing, China.,State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Wei Zhang
- State Key Laboratory for Quality Research in Chinese Medicines, Macau University of Science and TechnologyMacau, China
| | - Zun-Jian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education (MOE), China Pharmaceutical UniversityNanjing, China.,State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Feng-Guo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education (MOE), China Pharmaceutical UniversityNanjing, China.,State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| |
Collapse
|
20
|
Yao Y, Zhang P, Wang J, Chen J, Wang Y, Huang Y, Zhang Z, Xu F. Dissecting Target Toxic Tissue and Tissue Specific Responses of Irinotecan in Rats Using Metabolomics Approach. Front Pharmacol 2017; 8:122. [PMID: 28344557 PMCID: PMC5344918 DOI: 10.3389/fphar.2017.00122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 02/27/2017] [Indexed: 12/26/2022] Open
Abstract
As an anticancer agent, irinotecan (CPT-11) has been widely applied in clinical, especially in the treatment of colorectal cancer. However, its clinical use has long been limited by the side effects and potential tissue toxicity. To discriminate the target toxic tissues and dissect the specific response of target tissues after CPT-11 administration in rats, untargeted metabolomic study was conducted. First, differential metabolites between CPT-11 treated group and control group in each tissue were screened out. Then, based on fold changes of these differential metabolites, principal component analysis and hierarchical cluster analysis were performed to visualize the degree and specificity of the influences of CPT-11 on the metabolic profiles of nine tissues. Using this step-wise method, ileum, jejunum, and liver were finally recognized as target toxic tissues. Furthermore, tissue specific responses of liver, ileum, and jejunum to CPT-11 were dissected and specific differential metabolites were screened out. Perturbations in Krebs cycle, amino acid, purine and bile acid metabolism were observed in target toxic tissues. In conclusion, our study put forward a new approach to dissect target toxic tissues and tissue specific responses of CPT-11 using metabolomics.
Collapse
Affiliation(s)
- Yiran Yao
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Jing Wang
- School of Pharmacy, Shanxi University of Chinese Medicine Xianyang, China
| | - Jiaqing Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Yong Wang
- Jiangsu Institute for Food and Drug Control Nanjing, China
| | - Yin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| |
Collapse
|
21
|
Gonzalez FJ, Jiang C, Patterson AD. An Intestinal Microbiota-Farnesoid X Receptor Axis Modulates Metabolic Disease. Gastroenterology 2016; 151:845-859. [PMID: 27639801 PMCID: PMC5159222 DOI: 10.1053/j.gastro.2016.08.057] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/02/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023]
Abstract
The gut microbiota is associated with metabolic diseases including obesity, insulin resistance, and nonalcoholic fatty liver disease, as shown by correlative studies and by transplant of microbiota from obese humans and mice into germ-free mice. Modification of the microbiota by treatment of high-fat diet (HFD)-fed mice with tempol or antibiotics resulted in decreased adverse metabolic phenotypes. This was owing to lower levels of the genera Lactobacillus and decreased bile salt hydrolase (BSH) activity. The decreased BSH resulted in increased levels of tauro-β-muricholic acid (MCA), a substrate of BSH and a potent farnesoid X receptor (FXR) antagonist. Mice lacking expression of FXR in the intestine were resistant to HFD-induced obesity, insulin resistance, and nonalcoholic fatty liver disease, thus confirming that intestinal FXR is involved in the potentiation of metabolic disease. A potent intestinal FXR antagonist, glycine-β-MCA (Gly-MCA), which is resistant to BSH, was developed, which, when administered to HFD-treated mice, mimics the effect of the altered microbiota on HFD-induced metabolic disease. Gly-MCA had similar effects on genetically obese leptin-deficient mice. The decrease in adverse metabolic phenotype by tempol, antibiotics, and Gly-MCA was caused by decreased serum ceramides. Mice lacking FXR in the intestine also have lower serum ceramide levels, and are resistant to HFD-induced metabolic disease, and this was reversed by injection of C16:0 ceramide. In mouse ileum, because of the presence of endogenous FXR agonists produced in the liver, FXR target genes involved in ceramide synthesis are activated and when Gly-MCA is administered they are repressed, which likely accounts for the decrease in serum ceramides. These studies show that ceramides produced in the ileum under control of FXR influence metabolic diseases.
Collapse
Affiliation(s)
- Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, P. R. China
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802
| |
Collapse
|