1
|
Leung HKM, Lo EKK, Chen C, Zhang F, Felicianna, Ismaiah MJ, El-Nezami H. Probiotic Mixture Attenuates Colorectal Tumorigenesis in Murine AOM/DSS Model by Suppressing STAT3, Inducing Apoptotic p53 and Modulating Gut Microbiota. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10405-1. [PMID: 39641861 DOI: 10.1007/s12602-024-10405-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide. The standard CRC chemo drug, 5-Fluorouracil (5-FU), has a poor response rate and chemoresistance, prompting the need for a more effective and affordable treatment. In this study, we aimed to evaluate whether Prohep, a novel probiotic mixture, would alleviate azoxymethane/dextran sodium sulfate (AOM/DSS)-induced colorectal tumorigenesis and enhance 5-FU efficacy and its mechanism. Our results suggested that Prohep showed stronger anti-tumorigenesis effects than 5-FU alone or when combined in the AOM/DSS model. Prohep significantly reduced the total tumor count, total tumor size, caecum weight, colonic crypt depth, colonic inflammation, and collagen fibrosis. Prohep downregulated pro-inflammatory TNF-α and proliferative p-STAT3 and upregulated apoptotic p53. Metagenomics analysis indicated that Prohep-enriched Helicobacter ganmani, Desulfovibrio porci, Helicobacter hepaticus, and Candidatus Borkfalkia ceftriaxoniphila were inversely correlated to the total tumor count. In addition, Prohep-enriched Prevotella sp. PTAC and Desulfovibrio porci were negatively correlated to AOM/DSS enriched bacteria, while forming a co-existing community with other beneficial bacteria. From KEGG analysis, Prohep downregulated CRC-related pathways and enhanced pathways related to metabolites suppressing CRC like menaquinone, tetrapyrrole, aminolevulinic acid, and tetrahydrofolate. From Metacyc analysis, Prohep downregulated CRC-related peptidoglycan, LPS, and uric acid biosynthesis, and conversion. Prohep elevated the biosynthesis of the beneficial L-lysine, lipoic acid, pyrimidine, and palmitate. Prohep also elevated metabolic pathways related to energy utilization of lactic acid-producing bacteria (LAB) and acetate producers. Similarly, fecal acetate concentration was upregulated by Prohep. To sum up, Prohep demonstrated exceptional anti-tumorigenesis effects in the AOM/DSS model, which revealed its potential to develop into a novel CRC therapeutic in the future.
Collapse
Affiliation(s)
- Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Marsena Jasiel Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong, 999077, China.
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, 70211, Kuopio, Finland.
| |
Collapse
|
2
|
Zhong S, Wang Z, Yang J, Jiang D, Wang K. Ferroptosis-related oxaliplatin resistance in multiple cancers: Potential roles and therapeutic Implications. Heliyon 2024; 10:e37613. [PMID: 39309838 PMCID: PMC11414570 DOI: 10.1016/j.heliyon.2024.e37613] [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: 08/23/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Oxaliplatin (OXA)-based therapy is effective in the treatment of multiple cancers. However, primary or acquired OXA resistance remains an emerging challenge for its clinical application. Ferroptosis is an iron-dependent mode of cell death that has been demonstrated to play an essential role in the chemoresistance of many drugs, including OXA. In particular, dysregulation of SLC7A11-GPX4, one of the major antioxidant systems of ferroptosis, was found in the OXA resistance of colorectal cancer (CRC) and hepatocellular carcinoma (HCC). In addition, Nrf2, the upstream regulator of GPX4 and many other antioxidant factors, is also involved in the OXA resistance of CRC and HCC. Inhibition of SLC7A11-GPX4 or Nrf2 by genetic deletion of pharmaceutical inhibition could significantly reverse OXA resistance. Long noncoding RNA (lncRNA) also participates in chemoresistance and ferroptosis of cancer cells. Specifically, LINC01134 promotes the recruitment of Nrf2 to the promoter of GPX4, thereby exerting transcriptional regulation of GPX4, which eventually increases the OXA sensitivity of HCC through upregulation of ferroptosis. On the other hand, a novel lncRNA DACT3-AS1 sensitizes gastric cancer cells to OXA through miR-181a-5p/sirtuin 1(SIRT1)-mediated ferroptosis. Therapies based on ferroptosis or a combination of OXA and ferroptosis enhancers could provide new therapeutic insights to overcome OXA resistance. In the present review, we present the current understanding of ferroptosis-related OXA resistance, highlight ferroptosis pathogenesis in OXA chemoresistance, and summarize available therapies that target OXA resistance by enhancing ferroptosis.
Collapse
Affiliation(s)
- Sijia Zhong
- Department of Gastrointestinal Surgery, the First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Zihan Wang
- Department of Oral Implantology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, Liaoning, 110122, China
| | - Jiaxi Yang
- Department of Gastrointestinal Surgery, the First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Di Jiang
- China University of Petroleum (East China), 66 Changjiang West Road, Qingdao, 266580, China
| | - Kewei Wang
- Department of Gastrointestinal Surgery, the First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| |
Collapse
|
3
|
Shakhpazyan NK, Mikhaleva LM, Bedzhanyan AL, Gioeva ZV, Mikhalev AI, Midiber KY, Pechnikova VV, Biryukov AE. Exploring the Role of the Gut Microbiota in Modulating Colorectal Cancer Immunity. Cells 2024; 13:1437. [PMID: 39273009 PMCID: PMC11394638 DOI: 10.3390/cells13171437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/26/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The gut microbiota plays an essential role in maintaining immune homeostasis and influencing the immune landscape within the tumor microenvironment. This review aims to elucidate the interactions between gut microbiota and tumor immune dynamics, with a focus on colorectal cancer (CRC). The review spans foundational concepts of immuno-microbial interplay, factors influencing microbiome composition, and evidence linking gut microbiota to cancer immunotherapy outcomes. Gut microbiota modulates anti-cancer immunity through several mechanisms, including enhancement of immune surveillance and modulation of inflammatory responses. Specific microbial species and their metabolic byproducts can significantly influence the efficacy of cancer immunotherapies. Furthermore, microbial diversity within the gut microbiota correlates with clinical outcomes in CRC, suggesting potential as a valuable biomarker for predicting response to immunotherapy. Conclusions: Understanding the relationship between gut microbiota and tumor immune responses offers potential for novel therapeutic strategies and biomarker development. The gut microbiota not only influences the natural history and treatment response of CRC but also serves as a critical modulator of immune homeostasis and anti-cancer activity. Further exploration into the microbiome's role could enhance the effectiveness of existing treatments and guide the development of new therapeutic modalities.
Collapse
Affiliation(s)
- Nikolay K Shakhpazyan
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Liudmila M Mikhaleva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Arkady L Bedzhanyan
- Department of Abdominal Surgery and Oncology II (Coloproctology and Uro-Gynecology), Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Zarina V Gioeva
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Alexander I Mikhalev
- Department of Hospital Surgery No. 2, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Konstantin Y Midiber
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
- Institute of Medicine, Peoples' Friendship University of Russia named after Patrice Lumumba, 6 Miklukho-Maklaya St., 117198 Moscow, Russia
| | - Valentina V Pechnikova
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| | - Andrey E Biryukov
- Avtsyn Research Institute of Human Morphology, Petrovsky National Research Center of Surgery, 119435 Moscow, Russia
| |
Collapse
|
4
|
Hussain N, Muccee F, Ashraf NM, Afsar T, Husain FM, Hamid A, Razak S. Comparative analysis of adhesion virulence protein FadA from gut-associated bacteria of colorectal cancer patients ( F. nucleatum) and healthy individuals ( E. cloacae). J Cancer 2024; 15:5492-5505. [PMID: 39308684 PMCID: PMC11414602 DOI: 10.7150/jca.98951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/18/2024] [Indexed: 09/25/2024] Open
Abstract
Background: Colorectal cancer (CRC) is a gastrointestinal disease linked with GIT microbial dysbiosis. The present study has targeted the comparative analysis of virulent factor FadA from gut-associated bacteria of CRC patients (F. nucleatum) and healthy individuals (E. cloacae). Methods: For this purpose, FadA protein sequences of fifteen strains of F. nucleatum and four strains of E. cloacae, were retrieved from the UniProt database. These sequences were analysed through VirulentPred, PSLpred, ProtParam, PFP-FunDSeqE, PROTEUS Structure Prediction Server, SWISS-MODEL, SAVES validation server, MEME suite 5.5.0, CAVER Web tool, Webserver VaxinPAD, HPEPDOCK and HDOCK servers. Results: FadA protein from F. nucleatum was found to exhibit significant differences as compared to E. nucleatum i.e. it exhibited helical configuration, cytoplasmic, periplasmic, outer-membrane and extracellular localisation, 2D structure comprising of 70-96% helix, 0% beta-sheet, 4-30% coils and 17-20 signal peptide residues, hydrophilicity, strongly acidic character and smaller number of antigenic epitopes. In contrast, FadA protein from E. nucleatum was found to have globular 3D configuration, cytoplasmic localisation, 2D structure (30-56% helix, 12-21% beta-sheet, 33-50% coils and 43 signal peptide residues), highly hydrophobic, slightly acidic and more number of antigenic epitopes. Docking analyses of virulent factors revealed their high binding affinities with previously reported inhibitory peptide and FAD-approved drug COX2. Conclusion: The wide range of differences not only provided us the reason for the role of FadA protein as a virulent factor in F. nucleatum but also might help us in designing virulent FadA protein inhibiting strategies including peptide-based vaccine adjuvants and drugs designing, modification of tunnels and catalytic pockets to reduce substrate binding and FAD approved drugs selection. Inhibition of this virulent factor in CRC patients' gut bacteria might result in oncogenesis regression and reduced death rate.
Collapse
Affiliation(s)
- Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain Campus, Al Ain 64141, Abu Dhabi, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi Campus, Abu Dhabi P. O. Box 112612, Abu Dhabi, United Arab Emirates
| | - Fatima Muccee
- School of Biochemistry and Biotechnology, University of Punjab, Lahore, 52254, Pakistan
| | - Naeem Mahmood Ashraf
- School of Biochemistry and Biotechnology, University of Punjab, Lahore, 52254, Pakistan
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Arslan Hamid
- University of Bonn, LIMES Institute (AG-Netea), Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
5
|
Li B, Wei Z, Wang Z, Xu F, Yang J, Lin B, Chen Y, Wenren H, Wu L, Guo X, Liu Y, Wei Y. Fusobacterium nucleatum induces oxaliplatin resistance by inhibiting ferroptosis through E-cadherin/β-catenin/GPX4 axis in colorectal cancer. Free Radic Biol Med 2024; 220:125-138. [PMID: 38657754 DOI: 10.1016/j.freeradbiomed.2024.04.226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024]
Abstract
Fusobacterium (F.) nucleatum is a carcinogenesis microbiota in colorectal cancer (CRC). Growing evidence shows that F. nucleatum contributes to chemoresistance. Ferroptosis is reported to restore the susceptibility of resistant cells to chemotherapy. However, the role of gut microbiota affecting ferroptosis in chemoresistance remains unclear. Here, we examined the CRC tissues of patients using 16S rRNA sequencing to investigate the possible connection between gut microbiota dysbiosis and the relapse of CRC. We found that a high abundance of F. nucleatum in CRC tissue is associated with relapse. We further demonstrated that F. nucleatum induced oxaliplatin resistance in vitro and in vivo. The transcriptome of an F. nucleatum-infected cell revealed ferroptosis was associated with F. nucleatum infection. We perform malondialdehyde, ferrous iron, and glutathione assays to verify the effect of F. nucleatum on ferroptosis under oxaliplatin treatment in vivo and in vitro. Mechanistically, F. nucleatum promoted oxaliplatin resistance by overexpressing GPX4 and then inhibiting ferroptosis. E-cadherin/β-catenin/TCF4 pathway conducted the GPX4 overexpression effect of F. nucleatum. The chromatin immuno-precipitation quantitative PCR (CHIP-qPCR) and dual-luciferase reporter assay showed that F. nucleatum promoted TCF4 binding with GPX4. We also determined the E-cadherin/β-catenin/TCF4/GPX4 axis related to tumor tissue F. nucleatum status and CRC relapse clinically. Here, we revealed the contribution of F. nucleatum to oxaliplatin resistance by inhibiting ferroptosis in CRC. Targeting F. nucleatum and ferroptosis will provide valuable insight into chemoresistance management and may improve outcomes for patients with CRC.
Collapse
Affiliation(s)
- Bowen Li
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China
| | - Zixian Wei
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiyue Wang
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China
| | - Fangqi Xu
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China
| | - Jinhua Yang
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China
| | - Baiqiang Lin
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Chen
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China
| | - Hubin Wenren
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China
| | - Lingli Wu
- Department of Cardiovascular Medicine, Beilun District People's Hospital, Ningbo, China
| | - Xiao Guo
- Department of Breast Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Yang Liu
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China.
| | - Yunwei Wei
- Department of Oncology and Laparoscopy Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo No.2 Hospital, Ningbo, China; Key Laboratory of Intestinal Microecology and Major Human Diseases in Ningbo, Ningbo, China.
| |
Collapse
|
6
|
Thapa D, Kumar V, Naik B, Kumar V, Gupta AK, Mohanta YK, Mishra B, Rustagi S. Harnessing probiotic foods: managing cancer through gut health. Food Sci Biotechnol 2024; 33:2141-2160. [PMID: 39130664 PMCID: PMC11315834 DOI: 10.1007/s10068-024-01638-5] [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: 04/26/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 08/13/2024] Open
Abstract
One of the greatest threats to global health is cancer. Probiotic foods have been shown to have therapeutic promise in the management of cancer, even though traditional treatments such as radiation therapy, chemotherapy, and surgery are still essential. The generation of anticarcinogenic compounds, immune system stimulation, and gut microbiota regulation are a few ways that probiotics when taken in sufficient quantities, might help health. The purpose of this review is to examine the therapeutic potential of probiotic foods in the management of cancer. Research suggests that certain strains of probiotics have anticancer effects by preventing the growth of cancer cells, triggering apoptosis, and reducing angiogenesis in new tumors. Probiotics have shown promise in mitigating treatment-related adverse effects, such as diarrhea, mucositis, and immunosuppression caused by chemotherapy, improving the general quality of life for cancer patients. However, there are several factors, such as patient-specific features, cancer subtype, and probiotic strain type and dosage, which affect how effective probiotic therapies are in managing cancer. More research is necessary to find the long-term safety and efficacy characteristics of probiotics as well as to clarify the best ways to incorporate them into current cancer treatment methods. Graphical abstract Graphical representation showing the role of probiotic foods in cancer management.
Collapse
Affiliation(s)
- Devika Thapa
- Department of Food Science and Technology, Graphic Era Deemed to be University, Clement Town, Dehradun, Uttarakhand 248002 India
| | - Vijay Kumar
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand 248140 India
| | - Bindu Naik
- Department of Food Science and Technology, Graphic Era Deemed to be University, Clement Town, Dehradun, Uttarakhand 248002 India
- School of Agriculture, Graphic Era Hill University, Dehradun, Uttarakhand India
| | - Vivek Kumar
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun, Uttarakhand 248140 India
| | - Arun Kumar Gupta
- Department of Food Science and Technology, Graphic Era Deemed to be University, Clement Town, Dehradun, Uttarakhand 248002 India
| | - Yugal Kishore Mohanta
- Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua, Ri-Bhoi, Meghalaya 793101 India
| | - Bishwambhar Mishra
- Department of Biotechnology, Chaitanya Bharathi Institute of Technology (CBIT), Gandipet, Hyderabad, Telangana 500075 India
| | - Sarvesh Rustagi
- Department of Food Technology, SALS, Uttaranchal University, Dehradun, 248007 Uttarakhand India
| |
Collapse
|
7
|
Yim HCH, El-Omar E, Wu JCY, Sung JJY, Lee YY. Recent advances in translating gut microbiota research into clinical practice at 12th Asian Pacific Topic Conference 2023. J Gastroenterol Hepatol 2024; 39:979-981. [PMID: 38629218 DOI: 10.1111/jgh.16565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/26/2024] [Indexed: 06/19/2024]
Affiliation(s)
- Howard Chi Ho Yim
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Emad El-Omar
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Justin Che-Yuen Wu
- Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Joseph Jao Yiu Sung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yeong Yeh Lee
- School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
8
|
Deng Y, Hou X, Wang H, Du H, Liu Y. Influence of Gut Microbiota-Mediated Immune Regulation on Response to Chemotherapy. Pharmaceuticals (Basel) 2024; 17:604. [PMID: 38794174 PMCID: PMC11123941 DOI: 10.3390/ph17050604] [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: 03/12/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
The involvement of the gut microbiota in anti-cancer treatment has gained increasing attention. Alterations to the structure and function of the gut bacteria are important factors in the development of cancer as well as the efficacy of chemotherapy. Recent studies have confirmed that the gut microbiota and related metabolites influence the pharmacological activity of chemotherapeutic agents through interactions with the immune system. This review aims to summarize the current knowledge of how malignant tumor and chemotherapy affect the gut microbiota, how the gut microbiota regulates host immune response, and how interactions between the gut microbiota and host immune response influence the efficacy of chemotherapy. Recent advances in strategies for increasing the efficiency of chemotherapy based on the gut microbiota are also described. Deciphering the complex homeostasis maintained by the gut microbiota and host immunity provides a solid scientific basis for bacterial intervention in chemotherapy.
Collapse
Affiliation(s)
- Yufei Deng
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; (Y.D.); (X.H.); (H.W.)
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Xiaoying Hou
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; (Y.D.); (X.H.); (H.W.)
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
| | - Haiping Wang
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; (Y.D.); (X.H.); (H.W.)
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
| | - Hongzhi Du
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yuchen Liu
- Wuhan Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; (Y.D.); (X.H.); (H.W.)
- Cancer Institute, School of Medicine, Jianghan University, Wuhan 430056, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Jianghan University, Wuhan 430056, China
| |
Collapse
|
9
|
Jin M, Fan Q, Shang F, Zhang T, Ogino S, Liu H. Fusobacteria alterations are associated with colorectal cancer liver metastasis and a poor prognosis. Oncol Lett 2024; 27:235. [PMID: 38596264 PMCID: PMC11003219 DOI: 10.3892/ol.2024.14368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/01/2024] [Indexed: 04/11/2024] Open
Abstract
Liver metastasis is a major cause of mortality in patients with advanced stages of colorectal cancer (CRC). The gut microbiota has been demonstrated to influence the progression of liver diseases, potentially providing novel perspectives for diagnosis, treatment and research. However, the gut microbial characteristics in CRC with liver metastasis (LM) and with no liver metastasis (NLM) have not yet been fully established. In the present study, high-throughput 16S RNA sequencing technology was employed, in order to examine the gut microbial richness and composition in patients with CRC with LM or NLM. A discovery cohort (cohort 2; LM=18; NLM=36) and a validation cohort (cohort 3; LM=13; NLM=41) were established using fresh feces. In addition, primary carcinoma tissue samples were also analyzed (LM=8 and NLM=10) as a supplementary discovery cohort (cohort 1). The findings of the present study indicated that the intestinal microbiota richness and diversity were increased in the LM group as compared to the NLM group. A significant difference was observed in species composition between the LM and NLM group. In the two discovery cohorts with two different samples, the dominant phyla were consistent, but varied at lower taxonomic levels. Phylum Fusobacteria presented consistent and significant enrichment in LM group in both discovery cohorts. Furthermore, with the application of a random forest model and receiver operator characteristic curve analysis, Fusobacteria was identified as a potential biomarker for LM. Moreover, Fusobacteria was also a poor prognosis factor for survival. Importantly, the findings were reconfirmed in the validation cohort. On the whole, the findings of the present study demonstrated that CRC with LM and NLM exhibit distinct gut microbiota characteristics. Fusobacteria detection thus has potential for use in predicting LM and a poor prognosis of patients with CRC.
Collapse
Affiliation(s)
- Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Qilin Fan
- Department of Gastroenterology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Fumei Shang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shuji Ogino
- Program in Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02212, USA
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
- Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
10
|
Hui B, Zhou C, Xu Y, Wang R, Dong Y, Zhou Y, Ding J, Zhang X, Xu J, Gu Y. Exosomes secreted by Fusobacterium nucleatum-infected colon cancer cells transmit resistance to oxaliplatin and 5-FU by delivering hsa_circ_0004085. J Nanobiotechnology 2024; 22:62. [PMID: 38360615 PMCID: PMC10867993 DOI: 10.1186/s12951-024-02331-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND A large number of Fusobacterium nucleatum (Fn) are present in colorectal cancer (CRC) tissues of patients who relapse after chemotherapy, and Fn has been reported to promote oxaliplatin and 5-FU chemoresistance in CRC. Pathogens such as bacteria and parasites stimulate exosome production in tumor cells, and the regulatory mechanism of exosomal circRNA in the transmission of oxaliplatin and 5-FU chemotherapy resistance in Fn-infected CRC remains unclear. METHODS Hsa_circ_0004085 was screened by second-generation sequencing of CRC tissues. The correlation between hsa_circ_0004085 and patient clinical response to oxaliplatin/5-FU was analyzed. Exosome tracing experiments and live imaging systems were used to test the effect of Fn infection in CRC on the distribution of hsa_circ_0004085. Colony formation, ER tracking analysis and immunofluorescence were carried out to verify the regulatory effect of exosomes produced by Fn-infected CRC cells on chemotherapeutic resistance and ER stress. RNA pulldown, LC-MS/MS analysis and RIP were used to explore the regulatory mechanism of downstream target genes by hsa_circ_0004085. RESULTS First, we screened out hsa_circ_0004085 with abnormally high expression in CRC clinical samples infected with Fn and found that patients with high expression of hsa_circ_0004085 in plasma had a poor clinical response to oxaliplatin/5-FU. Subsequently, the circular structure of hsa_circ_0004085 was identified. Fn infection promoted hsa_circ_0004085 formation by hnRNP L and packaged hsa_circ_0004085 into exosomes by hnRNP A1. Exosomes produced by Fn-infected CRC cells transferred hsa_circ_0004085 between cells and delivered oxaliplatin/5-FU resistance to recipient cells by relieving ER stress. Hsa_circ_0004085 enhanced the stability of GRP78 mRNA by binding to RRBP1 and promoted the nuclear translocation of ATF6p50 to relieve ER stress. CONCLUSIONS Plasma levels of hsa_circ_0004085 are increased in colon cancer patients with intracellular Fn and are associated with a poor response to oxaliplatin/5-FU. Fn infection promoted hsa_circ_0004085 formation by hnRNP L and packaged hsa_circ_0004085 into exosomes by hnRNP A1. Exosomes secreted by Fn-infected CRC cells deliver hsa_circ_0004085 between cells. Hsa_circ_0004085 relieves ER stress in recipient cells by regulating GRP78 and ATF6p50, thereby delivering resistance to oxaliplatin and 5-FU.
Collapse
Affiliation(s)
- Bingqing Hui
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenchen Zhou
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yetao Xu
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Wang
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuwen Dong
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yirui Zhou
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Ding
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Zhang
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jian Xu
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yanhong Gu
- Department of Oncology and Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Van Dingenen L, Segers C, Wouters S, Mysara M, Leys N, Kumar-Singh S, Malhotra-Kumar S, Van Houdt R. Dissecting the role of the gut microbiome and fecal microbiota transplantation in radio- and immunotherapy treatment of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1298264. [PMID: 38035338 PMCID: PMC10687483 DOI: 10.3389/fcimb.2023.1298264] [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: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and poses a major burden on the human health worldwide. At the moment, treatment of CRC consists of surgery in combination with (neo)adjuvant chemotherapy and/or radiotherapy. More recently, immune checkpoint blockers (ICBs) have also been approved for CRC treatment. In addition, recent studies have shown that radiotherapy and ICBs act synergistically, with radiotherapy stimulating the immune system that is activated by ICBs. However, both treatments are also associated with severe toxicity and efficacy issues, which can lead to temporary or permanent discontinuation of these treatment programs. There's growing evidence pointing to the gut microbiome playing a role in these issues. Some microorganisms seem to contribute to radiotherapy-associated toxicity and hinder ICB efficacy, while others seem to reduce radiotherapy-associated toxicity or enhance ICB efficacy. Consequently, fecal microbiota transplantation (FMT) has been applied to reduce radio- and immunotherapy-related toxicity and enhance their efficacies. Here, we have reviewed the currently available preclinical and clinical data in CRC treatment, with a focus on how the gut microbiome influences radio- and immunotherapy toxicity and efficacy and if these treatments could benefit from FMT.
Collapse
Affiliation(s)
- Lena Van Dingenen
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Charlotte Segers
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Shari Wouters
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Mohamed Mysara
- Bioinformatics Group, Center for Informatics Science, School of Information Technology and Computer Science, Nile University, Giza, Egypt
| | - Natalie Leys
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Rob Van Houdt
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| |
Collapse
|
12
|
Kalaei Z, Manafi-Farid R, Rashidi B, Kiani FK, Zarei A, Fathi M, Jadidi-Niaragh F. The Prognostic and therapeutic value and clinical implications of fibroblast activation protein-α as a novel biomarker in colorectal cancer. Cell Commun Signal 2023; 21:139. [PMID: 37316886 DOI: 10.1186/s12964-023-01151-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/28/2023] [Indexed: 06/16/2023] Open
Abstract
The identification of contributing factors leading to the development of Colorectal Cancer (CRC), as the third fatal malignancy, is crucial. Today, the tumor microenvironment has been shown to play a key role in CRC progression. Fibroblast-Activation Protein-α (FAP) is a type II transmembrane cell surface proteinase expressed on the surface of cancer-associated fibroblasts in tumor stroma. As an enzyme, FAP has di- and endoprolylpeptidase, endoprotease, and gelatinase/collagenase activities in the Tumor Microenvironment (TME). According to recent reports, FAP overexpression in CRC contributes to adverse clinical outcomes such as increased lymph node metastasis, tumor recurrence, and angiogenesis, as well as decreased overall survival. In this review, studies about the expression level of FAP and its associations with CRC patients' prognosis are reviewed. High expression levels of FAP and its association with clinicopathological factors have made as a potential target. In many studies, FAP has been evaluated as a therapeutic target and diagnostic factor into which the current review tries to provide a comprehensive insight. Video Abstract.
Collapse
Affiliation(s)
- Zahra Kalaei
- Department of Biology, Faculty of Natural Sciences, Tabriz University, Tabriz, Iran
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Fathi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Domingues C, Cabral C, Jarak I, Veiga F, Dourado M, Figueiras A. The Debate between the Human Microbiota and Immune System in Treating Aerodigestive and Digestive Tract Cancers: A Review. Vaccines (Basel) 2023; 11:vaccines11030492. [PMID: 36992076 DOI: 10.3390/vaccines11030492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023] Open
Abstract
The human microbiota comprises a group of microorganisms co-existing in the human body. Unbalanced microbiota homeostasis may impact metabolic and immune system regulation, shrinking the edge between health and disease. Recently, the microbiota has been considered a prominent extrinsic/intrinsic element of cancer development and a promising milestone in the modulation of conventional cancer treatments. Particularly, the oral cavity represents a yin-and-yang target site for microorganisms that can promote human health or contribute to oral cancer development, such as Fusobacterium nucleatum. Moreover, Helicobacter pylori has also been implicated in esophageal and stomach cancers, and decreased butyrate-producing bacteria, such as Lachnospiraceae spp. and Ruminococcaceae, have demonstrated a protective role in the development of colorectal cancer. Interestingly, prebiotics, e.g., polyphenols, probiotics (Faecalibacterium, Bifidobacterium, Lactobacillus, and Burkholderia), postbiotics (inosine, butyrate, and propionate), and innovative nanomedicines can modulate antitumor immunity, circumventing resistance to conventional treatments and could complement existing therapies. Therefore, this manuscript delivers a holistic perspective on the interaction between human microbiota and cancer development and treatment, particularly in aerodigestive and digestive cancers, focusing on applying prebiotics, probiotics, and nanomedicines to overcome some challenges in treating cancer.
Collapse
Affiliation(s)
- Cátia Domingues
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Cristiana Cabral
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ivana Jarak
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Veiga
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Marília Dourado
- Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Health Studies and Research of the University of Coimbra (CEISUC), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Studies and Development of Continuous and Palliative Care (CEDCCP), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Figueiras
- Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV-REQUIMTE, Laboratory of Drug Development and Technologies, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
14
|
Lin ES, Huang YH, Yang PC, Peng WF, Huang CY. Complexed Crystal Structure of the Dihydroorotase Domain of Human CAD Protein with the Anticancer Drug 5-Fluorouracil. Biomolecules 2023; 13:149. [PMID: 36671534 PMCID: PMC9856072 DOI: 10.3390/biom13010149] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Dihydroorotase (DHOase) is the third enzyme in the pathway used for the biosynthesis of pyrimidine nucleotides. In mammals, DHOase is active in a trifunctional enzyme, CAD, which also carries out the activities of carbamoyl phosphate synthetase and aspartate transcarbamoylase. Prior to this study, it was unknown whether the FDA-approved clinical drug 5-fluorouracil (5-FU), which is used as an anticancer therapy, could bind to the DHOase domain of human CAD (huDHOase). Here, we identified huDHOase as a new 5-FU binding protein, thereby extending the 5-FU interactome to this human enzyme. In order to investigate where 5-FU binds to huDHOase, we solved the complexed crystal structure at 1.97 Å (PDB ID 8GVZ). The structure of huDHOase complexed with malate was also determined for the sake of comparison (PDB ID 8GW0). These two nonsubstrate ligands were bound at the active site of huDHOase. It was previously established that the substrate N-carbamoyl-L-aspartate is either bound to or moves away from the active site, but it is the loop that is extended towards (loop-in mode) or moved away (loop-out mode) from the active site. DHOase also binds to nonsubstrate ligands via the loop-out mode. In contrast to the Escherichia coli DHOase model, our complexed structures revealed that huDHOase binds to either 5-FU or malate via the loop-in mode. We further characterized the binding of 5-FU to huDHOase using site-directed mutagenesis and the fluorescence quenching method. Considering the loop-in mode, the dynamic loop in huDHOase should be a suitable drug-targeting site for further designing inhibitors and clinical chemotherapies to suppress pyrimidine biosynthesis in cancer cell lines.
Collapse
Affiliation(s)
- En-Shyh Lin
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung City 403, Taiwan
| | - Yen-Hua Huang
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung City 403, Taiwan
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Po-Chun Yang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
| | - Wei-Feng Peng
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
- Department of Medicine, College of Medicine, Chung Shan Medical University, Taichung City 402, Taiwan
- Department of Pediatrics, National Taiwan University Children’s Hospital, Taipei 100, Taiwan
| | - Cheng-Yang Huang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung City 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung City 402, Taiwan
| |
Collapse
|
15
|
Yu C, Zhou Z, Liu B, Yao D, Huang Y, Wang P, Li Y. Investigation of trends in gut microbiome associated with colorectal cancer using machine learning. Front Oncol 2023; 13:1077922. [PMID: 36937384 PMCID: PMC10015000 DOI: 10.3389/fonc.2023.1077922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/16/2023] [Indexed: 03/06/2023] Open
Abstract
Background The rapid growth of publications on the gut microbiome and colorectal cancer (CRC) makes it feasible for text mining and bibliometric analysis. Methods Publications were retrieved from the Web of Science. Bioinformatics analysis was performed, and a machine learning-based Latent Dirichlet Allocation (LDA) model was used to identify the subfield research topics. Results A total of 5,696 publications related to the gut microbiome and CRC were retrieved from the Web of Science Core Collection from 2000 to 2022. China and the USA were the most productive countries. The top 25 references, institutions, and authors with the strongest citation bursts were identified. Abstracts from all 5,696 publications were extracted for a text mining analysis that identified the top 50 topics in this field with increasing interest. The colitis animal model, expression of cytokines, microbiome sequencing and 16s, microbiome composition and dysbiosis, and cell growth inhibition were increasingly noticed during the last two years. The 50 most intensively investigated topics were identified and further categorized into four clusters, including "microbiome sequencing and tumor," "microbiome compositions, interactions, and treatment," "microbiome molecular features and mechanisms," and "microbiome and metabolism." Conclusion This bibliometric analysis explores the historical research tendencies in the gut microbiome and CRC and identifies specific topics of increasing interest. The developmental trajectory, along with the noticeable research topics characterized by this analysis, will contribute to the future direction of research in CRC and its clinical translation.
Collapse
|
16
|
The crosstalk of the human microbiome in breast and colon cancer: A metabolomics analysis. Crit Rev Oncol Hematol 2022; 176:103757. [PMID: 35809795 DOI: 10.1016/j.critrevonc.2022.103757] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/28/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
The human microbiome's role in colon and breast cancer is described in this review. Understanding how the human microbiome and metabolomics interact with breast and colon cancer is the chief area of this study. First, the role of the gut and distal microbiome in breast and colon cancer is investigated, and the direct relationship between microbial dysbiosis and breast and colon cancer is highlighted. This work also focuses on the many metabolomic techniques used to locate prospective biomarkers, make an accurate diagnosis, and research new therapeutic targets for cancer treatment. This review clarifies the influence of anti-tumor medications on the microbiota and the proactive measures that can be taken to treat cancer using a variety of therapies, including radiotherapy, chemotherapy, next-generation biotherapeutics, gene-based therapy, integrated omics technology, and machine learning.
Collapse
|
17
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
18
|
Hu LF, Lan HR, Huang D, Li XM, Jin KT. Personalized Immunotherapy in Colorectal Cancers: Where Do We Stand? Front Oncol 2021; 11:769305. [PMID: 34888246 PMCID: PMC8649954 DOI: 10.3389/fonc.2021.769305] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer death in the world. Immunotherapy using monoclonal antibodies, immune-checkpoint inhibitors, adoptive cell therapy, and cancer vaccines has raised great hopes for treating poor prognosis metastatic CRCs that are resistant to the conventional therapies. However, high inter-tumor and intra-tumor heterogeneity hinder the success of immunotherapy in CRC. Patients with a similar tumor phenotype respond differently to the same immunotherapy regimen. Mutation-based classification, molecular subtyping, and immunoscoring of CRCs facilitated the multi-aspect grouping of CRC patients and improved immunotherapy. Personalized immunotherapy using tumor-specific neoantigens provides the opportunity to consider each patient as an independent group deserving of individualized immunotherapy. In the recent decade, the development of sequencing and multi-omics techniques has helped us classify patients more precisely. The expansion of such advanced techniques along with the neoantigen-based immunotherapy could herald a new era in treating heterogeneous tumors such as CRC. In this review article, we provided the latest findings in immunotherapy of CRC. We elaborated on the heterogeneity of CRC patients as a bottleneck of CRC immunotherapy and reviewed the latest advances in personalized immunotherapy to overcome CRC heterogeneity.
Collapse
Affiliation(s)
- Li-Feng Hu
- Department of Colorectal Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Dong Huang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xue-Min Li
- Department of Hepatobiliary Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|