1
|
Yang Q, Wang B, Zheng Q, Li H, Meng X, Zhou F, Zhang L. A Review of Gut Microbiota-Derived Metabolites in Tumor Progression and Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207366. [PMID: 36951547 PMCID: PMC10214247 DOI: 10.1002/advs.202207366] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/15/2023] [Indexed: 05/27/2023]
Abstract
Gut microbiota-derived metabolites are key hubs connecting the gut microbiome and cancer progression, primarily by remodeling the tumor microenvironment and regulating key signaling pathways in cancer cells and multiple immune cells. The use of microbial metabolites in radiotherapy and chemotherapy mitigates the severe side effects from treatment and improves the efficacy of treatment. Immunotherapy combined with microbial metabolites effectively activates the immune system to kill tumors and overcomes drug resistance. Consequently, various novel strategies have been developed to modulate microbial metabolites. Manipulation of genes involved in microbial metabolism using synthetic biology approaches directly affects levels of microbial metabolites, while fecal microbial transplantation and phage strategies affect levels of microbial metabolites by altering the composition of the microbiome. However, some microbial metabolites harbor paradoxical functions depending on the context (e.g., type of cancer). Furthermore, the metabolic effects of microorganisms on certain anticancer drugs such as irinotecan and gemcitabine, render the drugs ineffective or exacerbate their adverse effects. Therefore, a personalized and comprehensive consideration of the patient's condition is required when employing microbial metabolites to treat cancer. The purpose of this review is to summarize the correlation between gut microbiota-derived metabolites and cancer, and to provide fresh ideas for future scientific research.
Collapse
Affiliation(s)
- Qiqing Yang
- General SurgeryCancer CenterDepartment of Breast SurgeryZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)Hangzhou310058China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Bin Wang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Qinghui Zheng
- General SurgeryCancer CenterDepartment of Breast SurgeryZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)Hangzhou310058China
| | - Heyu Li
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
| | - Xuli Meng
- General SurgeryCancer CenterDepartment of Breast SurgeryZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)Hangzhou310058China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058China
- International Biomed‐X Research CenterSecond Affiliated Hospital of Zhejiang University School of MedicineZhejiang UniversityHangzhou310058China
- Center for Infection & Immunity of International Institutes of Medicine The Fourth Affiliated HospitalZhejiang University School of MedicineYiwu322000China
- Cancer CenterZhejiang UniversityHangzhou310058China
| |
Collapse
|
2
|
Molecular imaging of aberrant crypt foci in the human colon targeting glutathione S-transferase P1-1. Sci Rep 2017; 7:6536. [PMID: 28747791 PMCID: PMC5529364 DOI: 10.1038/s41598-017-06857-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/19/2017] [Indexed: 01/06/2023] Open
Abstract
Aberrant crypt foci (ACF), the earliest precursor lesion of colorectal cancers (CRCs), are a good surrogate marker for CRC risk stratification and chemoprevention. However, the conventional ACF detection method with dye-spraying by magnifying colonoscopy is labor- and skill-intensive. We sought to identify rat and human ACF using a fluorescent imaging technique that targets a molecule specific for ACF. We found that glutathione S-transferase (GST) P1-1 was overexpressed in ACF tissues in a screening experiment. We then synthesized the fluorogenic probe, DNAT-Me, which is fluorescently quenched but is activated by GSTP1-1. A CRC cell line incubated with DNAT-Me showed strong fluorescence in the cytosol. Fluorescence intensities correlated significantly with GST activities in cancer cell lines. When we sprayed DNAT-Me onto colorectal mucosa excised from azoxymethane-treated rats and surgically resected from CRC patients, ACF with strong fluorescent signals were clearly observed. The ACF number determined by postoperative DNAT-Me imaging was almost identical to that determined by preoperative methylene blue staining. The signal-to-noise ratio for ACF in DNAT-Me images was significantly higher than that in methylene blue staining. Thus, we sensitively visualized ACF on rat and human colorectal mucosa by using a GST-activated fluorogenic probe without dye-spraying and magnifying colonoscopy.
Collapse
|
3
|
Zhang J, Jiang H, Xie L, Hu J, Li L, Yang M, Cheng L, Liu B, Qian X. Antitumor effect of manumycin on colorectal cancer cells by increasing the reactive oxygen species production and blocking PI3K-AKT pathway. Onco Targets Ther 2016; 9:2885-95. [PMID: 27307747 PMCID: PMC4888730 DOI: 10.2147/ott.s102408] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Manumycin is a natural, well-tolerated microbial metabolite and is regarded as a farnesyltransferase inhibitor. Some data suggest that manumycin inhibits proliferation of diverse cancer cells through various pathways. However, the antitumor effect of manumycin on colorectal cancer (CRC) remains unknown. In the present study, we investigated the antitumor effect of manumycin on CRC in vitro and in vivo. The results of cell viability assay revealed that the proliferation of the CRC cells was significantly inhibited by manumycin. Moreover, cell apoptosis induced by manumycin was also found in a time- and dose-dependent manner. Interestingly, treatment of the CRC cells with manumycin resulted in increased generation of reactive oxygen species. Subsequently, manumycin also decreased the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and AKT, as well as the expression of caspase-9 and poly(ADP-ribose) polymerase (PARP) in a time-dependent manner. In addition, we found that N-acetyl-l-cysteine (NAC) attenuated the effect of manumycin on the PI3K-AKT pathway, and wortmannin reduced the effect of manumycin on caspase-9 and PARP expression. More importantly, the anticancer effect of manumycin was also observed in established tumor xenografts. Taken together, these findings supported the potential application of manumycin against colorectal carcinoma.
Collapse
Affiliation(s)
- Jingyu Zhang
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hua Jiang
- Department of Oncology, Affiliated Changzhou No 2 People's Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Li Xie
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jing Hu
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Li Li
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Mi Yang
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lei Cheng
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Baorui Liu
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaoping Qian
- Department of the Comprehensive Cancer Center, Affiliated Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
4
|
Kim KH, Chae JI, Oh H, Cho JH, Lee RH, Yoon G, Cho SS, Cho YS, Lee MH, Liu K, Lee HJ, Shim JH. Manumycin A induces apoptosis in malignant pleural mesothelioma through regulation of Sp1 and activation of the mitochondria-related apoptotic pathway. Oncol Rep 2016; 36:117-24. [PMID: 27176604 DOI: 10.3892/or.2016.4801] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/11/2016] [Indexed: 11/06/2022] Open
Abstract
Manumycin A (Manu A) is a natural product isolated from Streptomyces parvulus and has been reported to have anti-carcinogenic and anti-biotic properties. However, neither its molecular mechanism nor its molecular targets are well understood. Thus, the aim of the present study was to explore the possibility that Manu A has cancer preventive and chemotherapeutic effects on malignant pleural mesothelioma (MPM) through regulation of Sp1 and induction of mitochondrial cell death pathway. Manu A inhibited the cell viability of MSTO-211H and H28 cells in a concentration‑dependent manner as determined by MTS assay. IC50 values were calculated as 8.3 and 4.3 µM in the MSTO-311H and H28 cells following 48 h incubation, respectively. Manu A induced a significant increase in apoptotic indices as shown by DAPI staining, Annexin V assay, multi-caspase activity and mitochondrial membrane potential assay. The downregulation of Sp1 mRNA and protein expression by Manu A led to apoptosis by suppressing Sp1-regulated proteins (cyclin D1, Mcl-1 and survivin). Manu A decreased the protein levels of BID, Bcl-xL and PARP while it increased Bax levels. Manu A caused depolarization of the mitochondrial membrane with induction of CHOP, DR4 and DR5. Our results demonstrated that Manu A exerted anticancer effects by inducing apoptosis via inhibition of the Sp1-related signaling pathway in human MPM.
Collapse
Affiliation(s)
- Ka Hwi Kim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 651-756, Republic of Korea
| | - Hana Oh
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Jin Hyoung Cho
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 651-756, Republic of Korea
| | - Ra-Ham Lee
- Department of Dental Pharmacology, School of Dentistry and Institute of Oral Bioscience, BK21 Plus, Chonbuk National University, Jeonju 651-756, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| | - Young-Sik Cho
- College of Pharmacy, Keimyung University, Dalseo-gu, Daegu 704-701, Republic of Korea
| | - Mee-Hyun Lee
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450001, P.R. China
| | - Kangdong Liu
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan 450001, P.R. China
| | - Hyun-Jeong Lee
- Division of Animal Genomics and Bioinformatics, National Institute of Animal Science, Rural Development Administration, Suwon 441-706, Republic of Korea
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 534-729, Republic of Korea
| |
Collapse
|
5
|
Caiazza F, Elliott L, Fennelly D, Sheahan K, Doherty GA, Ryan EJ. Targeting EGFR in metastatic colorectal cancer beyond the limitations of KRAS status: alternative biomarkers and therapeutic strategies. Biomark Med 2016; 9:363-75. [PMID: 25808440 DOI: 10.2217/bmm.15.5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Patients with metastatic colorectal cancer have a very poor prognosis. Incorporation of targeted molecular therapies, such as the anti-EGFR receptor monoclonal antibodies cetuximab and panitumumab, into treatment regimens has improved outcomes for patients with wild-type RAS tumors. Yet, response rates remain low and overall survival times are short. Increased understanding of oncogenic signaling pathways within the tumor, and how these are regulated by the inflammatory tumor microenvironment, is a priority to facilitate the development of biomarkers to better guide the use of existing therapies and to develop new ones. Here, we review recent preclinical and clinical progress in the development of biomarkers for predicting response to anti-EGFR therapy in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Francesco Caiazza
- Centre for Colorectal Disease, St. Vincent's University Hospital, Elm Park, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|